Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
FASEB J ; 37(12): e23292, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37971407

RESUMO

Immunotoxins (ITs) target cancer cells via antibody binding to surface antigens followed by internalization and toxin-mediated inhibition of protein synthesis. The fate of cells responding to IT treatment depends on the amount and stability of specific pro-apoptotic and pro-survival proteins. When treated with a pseudomonas exotoxin-based immunotoxin (HB21PE40), the triple-negative breast cancer (TNBC) cell line MDA-MB-468 displayed a notable resistance to toxin-mediated killing compared to the epidermoid carcinoma cell line, A431, despite succumbing to the same level of protein synthesis inhibition. In a combination screen of ~1912 clinically relevant and mechanistically annotated compounds, we identified several agents that greatly enhanced IT-mediated killing of MDA-MB-468 cells while exhibiting only a modest enhancement for A431 cells. Of interest, two Smac mimetics, birinapant and SM164, exhibited this kind of differential enhancement. To investigate the basis for this, we probed cells for the presence of inhibitor of apoptosis (IAP) proteins and monitored their stability after the addition of immunotoxin. We found that high levels of IAPs inhibited immunotoxin-mediated cell death. Further, TNFα levels were not relevant for the combination's efficacy. In tumor xenograft studies, combinations of immunotoxin and birinapant caused complete regressions in MDA-MB-468tumor-bearing mice but not in mice with A431 tumors. We propose that IAPs constitute a barrier to immunotoxin efficacy which can be overcome with combination treatments that include Smac mimetics.


Assuntos
Imunotoxinas , Neoplasias , Humanos , Animais , Camundongos , Proteínas Inibidoras de Apoptose/metabolismo , Imunotoxinas/farmacologia , Linhagem Celular Tumoral , Dipeptídeos/farmacologia , Apoptose
2.
Biomed Pharmacother ; 157: 114047, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36459711

RESUMO

The monoclonal antibody '40H3' binds to EGFRvIII and to full-length EGFR when it is overexpressed on cancer cells. To generate candidate cytotoxic antibody-drug conjugates (ADCs), 40H3 was modified by the addition of small molecular weight payloads that included two tubulin-modifying agents, two topoisomerase inhibitors and a pyrrolobenzodiazepine (PBD) dimer. Conjugates retained antigen binding activity comparable to the unmodified 40H3 antibody. The cytotoxicity of five distinct ADCs was evaluated on a variety of EGFR-expressing cells including three triple negative breast cancer (TNBC) lines. Generally, the 40H3 conjugate with the PBD dimer (40H3-Tesirine) was the most active killing agent. The killing of EGFR-positive cells by 40H3-Tesirine correlated with the number of surface binding sites for 40H3. However, bystander killing was also evident in experiments with antigen-negative cells. In vivo tumor xenograft experiments were conducted on two TNBC tumor lines. Three treatments with the 40H3-Tesirine ADC at 1 mg/kg were sufficient to achieve complete remissions without evidence of mouse toxicity. Data support the development of ADCs derived from the 40H3 antibody for the treatment of cancers that express EGFRvIII or high levels of EGFR.


Assuntos
Antineoplásicos , Imunoconjugados , Neoplasias , Humanos , Camundongos , Animais , Imunoconjugados/farmacologia , Imunoconjugados/uso terapêutico , Imunoconjugados/química , Ensaios Antitumorais Modelo de Xenoenxerto , Linhagem Celular Tumoral , Antineoplásicos/uso terapêutico , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Neoplasias/tratamento farmacológico , Receptores ErbB
3.
Front Immunol ; 13: 954992, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36341428

RESUMO

The tumor microenvironment (TME) influences tumor growth, metastatic spread and response to treatment. Often immunosuppression, mediated by the TME, impairs a beneficial response. The complexity of the tumor composition challenges our abilities to design new and more effective therapies. Going forward we will need to 'manage' the content and or functionality of the TME to improve treatment outcomes. Currently, several different kinds of treatments are available to patients with cancer: there are the traditional approaches of chemotherapy, radiation and surgery; there are targeted agents that inhibit kinases associated with oncogenic pathways; there are monoclonal antibodies that target surface antigens often delivering toxic payloads or cells and finally there are antibodies and biologics that seek to overcome the immunosuppression caused by elements within the TME. How each of these therapies interact with the TME is currently under intense and widespread investigation. In this review we describe how the TME and its immunosuppressive components can influence both tumor progression and response to treatment focusing on three particular tumor types, classic Hodgkin Lymphoma (cHL), Pancreatic Ductal Adenocarcinoma (PDAC) and Glioblastoma Multiforme (GBM). And, finally, we offer five approaches to manipulate or manage the TME to improve outcomes for cancer patients.


Assuntos
Antineoplásicos Imunológicos , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Microambiente Tumoral , Neoplasias Pancreáticas/patologia , Carcinoma Ductal Pancreático/patologia , Imunoterapia , Antineoplásicos Imunológicos/uso terapêutico
4.
Biomolecules ; 10(9)2020 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-32957689

RESUMO

Cancer cells frequently upregulate surface receptors that promote growth and survival. These receptors constitute valid targets for intervention. One strategy involves the delivery of toxic payloads with the goal of killing those cancer cells with high receptor levels. Delivery can be accomplished by attaching a toxic payload to either a receptor-binding antibody or a receptor-binding ligand. Generally, the cell-binding domain of the toxin is replaced with a ligand or antibody that dictates a new binding specificity. The advantage of this "immunotoxin" approach lies in the potency of these chimeric molecules for killing cancer cells. However, receptor expression on normal tissue represents a significant obstacle to therapeutic intervention.


Assuntos
Anticorpos Monoclonais/imunologia , Imunotoxinas/imunologia , Neoplasias/imunologia , Receptores de Superfície Celular/imunologia , Toxinas Biológicas/imunologia , Animais , Anticorpos Monoclonais/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/imunologia , Humanos , Imunotoxinas/metabolismo , Imunotoxinas/uso terapêutico , Ligantes , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Ligação Proteica , Receptores de Superfície Celular/antagonistas & inibidores , Receptores de Superfície Celular/metabolismo , Toxinas Biológicas/metabolismo
5.
Antib Ther ; 2(1): 1-10, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30801054

RESUMO

BACKGROUND: A feature of many B-cell tumors is a surface-expressed immunoglobulin (sIg). The complementarity determiningregions (CDRs)of the sIg, termed the 'idiotype', are unique to each tumor. We report on a phage selection strategy to generate anti-idiotype therapeutics that react with sIg CDR3 sequences: the MEC1 B-cell tumor line was used as proof of concept. METHODS: To create a mimetic of the MEC1 idiotype, CDR3 sequences from heavy and light chains of the sIg were grafted into a scFv framework scaffold. Using the Tomlinson I phage library of human scFvs, we enriched for binders to MEC1 CDR3 sequences over unrelated CDR3 sequences. RESULTS: By ELISA we identified 10 binder phage. Of these, five were sequenced, found to be unique and characterized further. By flow cytometry each of the five phage bound to MEC1 cells, albeit with different patterns of reactivity. To establish specificity of binding and utility, the scFv sequences from two of these binders (phage 1, and 7) were converted into antibody-toxin fusion proteins (immunotoxins) and also cloned into a human IgG1 expression vector. Binder-1 and -7 immunotoxins exhibited specific killing of MEC1 cells with little toxicity for non-target B-cell lines. The full-length antibody recreated from the binder-1 scFv so exhibited specific binding. CONCLUSION: Our results establish the utility of using engrafted CDR3 sequences for selecting phage that recognize the idiotype of B-cell tumors.

6.
JCI Insight ; 4(5)2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30720466

RESUMO

The routes by which antibody-based therapeutics reach malignant cells are poorly defined. Tofacitinib, an FDA-approved JAK inhibitor, reduced tumor-associated inflammatory cells and allowed increased delivery of antibody-based agents to malignant cells. Alone, tofacitinib exhibited no antitumor activity, but combinations with immunotoxins or an antibody-drug conjugate resulted in increased antitumor responses. Quantification using flow cytometry revealed that antibody-based agents accumulated in malignant cells at higher percentages following tofacitinib treatment. Profiling of tofacitinib-treated tumor-bearing mice indicated that cytokine transcripts and various proteins involved in chemotaxis were reduced compared with vehicle-treated mice. Histological analysis revealed significant changes to the composition of the tumor microenvironment, with reductions in monocytes, macrophages, and neutrophils. Tumor-associated inflammatory cells contributed to non-target uptake of antibody-based therapeutics, with mice treated with tofacitinib showing decreased accumulation of therapeutics in intratumoral inflammatory cells and increased delivery to malignant cells. The present findings serve as a rationale for conducting trials where short-term treatments with tofacitinib could be administered in combination with antibody-based therapies.


Assuntos
Anticorpos/farmacologia , Imunoconjugados/uso terapêutico , Neoplasias/tratamento farmacológico , Piperidinas/farmacologia , Pirimidinas/farmacologia , Pirróis/farmacologia , Animais , Anticorpos/uso terapêutico , Arginase , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Imunoterapia , Imunotoxinas , Macrófagos , Camundongos , Camundongos Nus , Monócitos , Neutrófilos , Piperidinas/uso terapêutico , Pirimidinas/uso terapêutico , Pirróis/uso terapêutico , RNA Mensageiro/metabolismo , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Antib Ther ; 2(4): 88-98, 2019 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-31934685

RESUMO

BACKGROUND: The dysregulation of epidermal growth factor receptor (EGFR) has been implicated in the oncogenesis of various malignancies including glioblastoma and some epithelial cancers. Oncogenesis occurs from the overexpression of EGFR, often linked to gene amplification or receptor mutagenesis. The 287-302 loop in the extracellular domain is exposed completely on EGFR variant III (EGFRvIII), partially exposed on some cancers but cryptic on normal cells. We report on the generation of antibodies to this loop. METHODS: The 286-303 peptide was coupled chemically to keyhole limpet hemocyanin. After immunizations, sera were assayed for reactivity to the peptide. Mice with high titers were used for hybridoma production. Purified antibodies were isolated from hybridoma supernatants, while V regions were cloned and sequenced. Receptor binding was characterized using enzyme-linked immunosorbent assay and flow cytometry. A recombinant immunotoxin was generated from the 40H3 antibody and its cytotoxic activity characterized on relevant cancer cell lines. RESULTS: Seven monoclonal antibodies were generated to the 287-302 loop and characterized further. Each one reacted with EGFRvIII but not wild-type EGFR. Based on reactivity with the immunizing peptide, antibodies were mapped to one of three subgroups. One antibody, 40H3, also exhibited binding to MDA-MB-468 and A431 cells but not to non-cancerous WI-38 cells. Because of its unusual binding characteristics, a recombinant immunotoxin was generated from 40H3, which proved to be cytotoxic to MDA-MB-468, A431 and F98npEGFRvIII expressing cells. CONCLUSIONS: Immunization with a peptide corresponding to a cryptic epitope from EGFR can produce tumor cell-binding antibodies. The 40H3 antibody was engineered as a cytotoxic recombinant immunotoxin and could be further developed as a therapeutic agent.

8.
PLoS One ; 11(8): e0161415, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27556570

RESUMO

The intersection of small molecular weight drugs and antibody-based therapeutics is rarely studied in large scale. Both types of agents are currently part of the cancer armamentarium. However, very little is known about how to combine them in optimal ways. Immunotoxins are antibody-toxin gene fusion proteins engineered to target cancer cells via antibody binding to surface antigens. For fusion proteins derived from Pseudomonas exotoxin (PE), potency relies on the enzymatic domain of the toxin which catalyzes the ADP-ribosylation of EF2 causing inhibition of protein synthesis leading to cell death. Candidate immunotoxins have demonstrated clear value in clinical trials but generally have not been curative as single agents. Therefore we undertook three screens to discover effective combinations that could act synergistically. From the MIPE-3 library of compounds we identified various enhancers of immunotoxin action and at least one major class of inhibitor. Follow-up experiments confirmed the screening data and suggested that immunotoxins when administered with everolimus or nilotinib exhibit favorable combinatory activity and would be candidates for preclinical development. Mechanistic studies revealed that everolimus-immunotoxin combinations acted synergistically on elements of the protein synthetic machinery, including S61 kinase and 4E-BP1 of the mTORC1 pathway. Conversely, PARP inhibitors antagonized immunotoxins and also blocked the toxicity due to native ADP-ribosylating toxins. Thus, our goal of investigating a chemical library was justified based on the identification of several approved compounds that could be developed preclinically as 'enhancers' and at least one class of mitigator to be avoided.


Assuntos
Anticorpos Monoclonais , Descoberta de Drogas , Ensaios de Seleção de Medicamentos Antitumorais , Proteínas Recombinantes de Fusão/farmacologia , Toxinas Biológicas , Animais , Anticorpos Monoclonais/genética , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Descoberta de Drogas/métodos , Exotoxinas/genética , Feminino , Humanos , Imunotoxinas/genética , Imunotoxinas/farmacologia , Camundongos , Inibidores de Poli(ADP-Ribose) Polimerases/farmacologia , Proteínas Recombinantes de Fusão/genética , Bibliotecas de Moléculas Pequenas , Toxinas Biológicas/genética , Ensaios Antitumorais Modelo de Xenoenxerto
9.
J Natl Cancer Inst ; 108(8)2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27075852

RESUMO

BACKGROUND: Triple-negative breast cancers (TNBCs) are typically more aggressive and result in poorer outcomes than other breast cancers because treatment options are limited due to lack of hormone receptors or amplified human epidermal growth factor receptor 2 (HER2). Many TNBCs overexpress the epidermal growth factor receptor (EGFR) or manifest amplification of theEGFRgene, supporting EGFR as a therapeutic target. While EGFR-directed small molecule inhibitors have shown limited effectiveness in clinical settings, use of EGFR as a mechanism of delivering enzymatic cytotoxins to TNBC has not been demonstrated. METHODS: Using the single-chain variable fragment (scFv) of the 806 antibody that binds only cells with overexpressed, misfolded, or mutant variants of the EGFR, a recombinant immunotoxin was engineered through gene fusion withPseudomonas aeruginosaExotoxin A (806-PE38). The potency of 806-PE38 on reducing TNBC cell growth in vitro and in xenograft models (n ≥ 6) was examined for six TNBC cell lines. All statistical tests were two-sided. RESULTS: 806-PE38 statistically significantly reduced the viability of all tested TNBC lines, with IC50values below 10 ng/mL for three of six cell lines, while not affecting cells with wild-type EGFR (IC50>300 ng/mL). Systemic treatments with 806-PE38 vs vehicle resulted in statistically significantly reduced tumor burdens (806-PE38 mean = 128 mm(3)[SD = 46 mm(3)] vs vehicle mean = 749 mm(3)[SD = 395 mm(3)], P = .001) and increased median survival (806-PE38 median = 82 days vs vehicle median = 50 days,P= .01) in a MDA-MB-468 TNBC mouse xenograft. Deletion of the catalytic residue eliminated both cytotoxic activity in vitro and the reduction in tumor burden and survival (P= .52). CONCLUSIONS: These data support the further development of the 806-PE38 immunotoxin as a therapeutic agent for the treatment of patients with EGFR-positive TNBC. Follow-up experiments with combination therapies will be attempted to achieve full remissions.


Assuntos
ADP Ribose Transferases/uso terapêutico , Toxinas Bacterianas/uso terapêutico , Receptores ErbB/imunologia , Exotoxinas/uso terapêutico , Fatores Imunológicos/uso terapêutico , Imunotoxinas/uso terapêutico , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/imunologia , Fatores de Virulência/uso terapêutico , ADP Ribose Transferases/farmacologia , Animais , Toxinas Bacterianas/farmacologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Portadores de Fármacos , Epitopos/imunologia , Receptores ErbB/genética , Exotoxinas/farmacologia , Feminino , Humanos , Fatores Imunológicos/farmacologia , Imunotoxinas/farmacologia , Camundongos , Camundongos Nus , Terapia de Alvo Molecular , Transplante de Neoplasias , Proteínas Recombinantes/uso terapêutico , Taxa de Sobrevida , Neoplasias de Mama Triplo Negativas/patologia , Carga Tumoral/efeitos dos fármacos , Fatores de Virulência/farmacologia
10.
J Biomol Screen ; 21(9): 942-55, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27112173

RESUMO

Synthetic lethal screens are used to discover new combination treatments for cancer. In traditional high-throughput synthetic lethal screens, compounds are tested at a single dose, and hit selection is based on threshold activity values from the variance of the efficacy of the compounds tested. The limitation of the single-dose screening for synthetic lethal screens is that it does not allow for the robust detection of differential activities from compound collections with a broad range of potencies and efficacies. There is therefore a need to develop screening approaches that enable the identification of compounds with synthetic lethal effects based on changes in both potency and efficacy. Here we describe the implementation of a dose response-based synthetic lethal screen to find drugs that enhance or mitigate the cytotoxic effect of an immunotoxin protein (HA22). We developed a data analysis framework for the selection of compounds with enhancing or mitigating cytotoxic activities based on the use of dose-response parameters. The data analysis framework includes an ensemble ranking approach that allows the use of multiple dose-response parameters in a nonparametric fashion. Quantitative high-throughput screening (HTS) enables the identification of compounds with synthetic lethal activity not identified by single-dose HTS.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Neoplasias/tratamento farmacológico , Bibliotecas de Moléculas Pequenas/farmacologia , Mutações Sintéticas Letais/genética , Toxinas Bacterianas/antagonistas & inibidores , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Exotoxinas/antagonistas & inibidores , Humanos , Neoplasias/genética , Bibliotecas de Moléculas Pequenas/isolamento & purificação , Bibliotecas de Moléculas Pequenas/uso terapêutico
11.
Protein Expr Purif ; 122: 56-63, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26923059

RESUMO

BCL-XL is a dominant inhibitor of apoptosis and a significant anti-cancer drug target. Endogenous BCL-XL is integral to the mitochondrial outer membrane (MOM). BCL-XL reconstituted in detergent-free lipid bilayer nanodiscs is anchored to the nanodisc lipid bilayer membrane by tight association of its C-terminal tail, while the N-terminal head retains the canonical structure determined for water-soluble, tail-truncated BCL-XL, with the surface groove solvent-exposed and available for BH3 ligand binding. To better understand the conformation and dynamics of this key region of BCL-XL we have developed methods for isolating the membrane-embedded C-terminal tail from its N-terminal head and for preparing protein suitable for structural and biochemical studies. Here, we outline the methods for sample preparation and characterization and describe previously unreported structural and dynamics features. We show that the C-terminal tail of BCL-XL forms a transmembrane α-helix that retains a significant degree of conformational dynamics. We also show that the presence of the intact C-terminus destabilizes the soluble state of the protein, and that the small fraction of soluble recombinant protein produced in Escherichia coli is susceptible to proteolytic degradation of C-terminal residues beyond M218. This finding impacts the numerous previous studies where recombinant soluble BCL-XL was presumed to be full-length. Nevertheless, the majority of recombinant BCL-XL produced in E. coli is insoluble and protected from proteolysis. This protein retains the complete C-terminal tail and can be reconstituted in lipid bilayers in a folded and active state.


Assuntos
Bicamadas Lipídicas/química , Proteína bcl-X/química , Sequência de Aminoácidos , Apoptose , Clonagem Molecular , Escherichia coli/genética , Humanos , Ressonância Magnética Nuclear Biomolecular , Conformação Proteica , Conformação Proteica em alfa-Hélice , Estabilidade Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Solubilidade , Proteína bcl-X/genética
12.
J Mol Biol ; 427(13): 2262-70, 2015 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-25731750

RESUMO

BCL-XL is an anti-apoptotic BCL-2 family protein found both in the cytosol and bound to intracellular membranes. Structural studies of BCL-XL have advanced by deleting its hydrophobic C-terminus and adding detergents to enhance solubility. However, since the C-terminus is essential for function and detergents strongly affect structure and activity, the molecular mechanisms controlling intracellular localization and cytoprotective activity are incompletely understood. Here we describe the conformations and ligand binding activities of water-soluble and membrane-bound BCL-XL, with its complete C-terminus, in detergent-free environments. We show that the C-terminus interacts with a conserved surface groove in the water-soluble state of the protein and inserts across the phospholipid bilayer in the membrane-bound state. Contrary to current models, membrane binding does not induce a conformational change in the soluble domain and both states bind a known ligand with affinities that are modulated by the specific state of the protein.


Assuntos
Membrana Celular/metabolismo , Proteína bcl-X/química , Proteína bcl-X/metabolismo , Proteínas Reguladoras de Apoptose/química , Proteínas Reguladoras de Apoptose/metabolismo , Sítios de Ligação , Calorimetria , Membrana Celular/química , Detergentes/química , Humanos , Bicamadas Lipídicas , Modelos Moleculares , Ressonância Magnética Nuclear Biomolecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fosfolipídeos/química , Conformação Proteica , Dobramento de Proteína , Multimerização Proteica , Estrutura Terciária de Proteína , Solubilidade
13.
Proc Natl Acad Sci U S A ; 112(10): E1135-42, 2015 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-25713356

RESUMO

Immunotoxins (antibody-toxin fusion proteins) target surface antigens on cancer cells and kill these cells via toxin-mediated inhibition of protein synthesis. To identify genes controlling this process, an RNAi whole-genome screen (∼ 22,000 genes at three siRNAs per gene) was conducted via monitoring the cytotoxicity of the mesothelin-directed immunotoxin SS1P. SS1P, a Pseudomonas exotoxin-based immunotoxin, was chosen because it is now in clinical trials and has produced objective tumor regressions in patients. High and low concentrations of SS1P were chosen to allow for the identification of both mitigators and sensitizers. As expected, silencing known essential genes in the immunotoxin pathway, such as mesothelin, furin, KDEL receptor 2, or members of the diphthamide pathway, protected cells. Of greater interest was the observation that many RNAi targets increased immunotoxin sensitivity, indicating that these gene products normally contribute to inefficiencies in the killing pathway. Of the top sensitizers, many genes encode proteins that locate to either the endoplasmic reticulum (ER) or Golgi and are annotated as part of the secretory system. Genes related to the ER-associated degradation system were not among high-ranking mitigator or sensitizer candidates. However, the p97 inhibitor eeyarestatin 1 enhanced immunotoxin killing. Our results highlight potential targets for chemical intervention that could increase immunotoxin killing of cancer cells and enhance our understanding of toxin trafficking.


Assuntos
Retículo Endoplasmático/metabolismo , Complexo de Golgi/metabolismo , Imunotoxinas/farmacologia , Interferência de RNA , Animais , Humanos
14.
Mol Cancer Ther ; 13(6): 1655-63, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24739394

RESUMO

Impaired apoptosis is often a key element in tumor development. Therefore, drugs mimicking prosurvival antagonists offer promise as cancer therapeutics. When ABT-737, a BH3-only mimetic, was added to KB3-1 human cervical adenocarcinoma cells, we noted an induction of an endoplasmic reticulum (ER) stress response and the dislocation of ER luminal proteins, including chaperones, to the cell cytosol. Furthermore, when immunotoxin (antibody-toxin chimeric molecule) and ABT-737 combinations were added to cells, there was enhanced toxin-mediated inhibition of protein synthesis, consistent with enhanced translocation of toxin to the cytosol. A similar enhancement was not seen with thapsigargin, suggesting that ER stress alone was not responsible for enhanced translocation. Cytosol preparations from ABT-737-treated but not from thapsigargin-treated cells revealed the presence of greater amounts of processed 37-kDa toxin fragment compared with the addition of immunotoxin alone. As early as 4 hours after the addition of ABT-737 and immunotoxin, there was release of mitochondrial cytochrome c and activation of caspase-3/7 indicating that the combination caused apoptotic cell death. These results were reflected in decreased cellular ATP levels that were noted with combinations of ABT-737 and immunotoxin but not with either agent alone or with combinations of thapsigargin and immunotoxin. We conclude that ABT-737 increases ER permeability, promoting the dislocation of toxin from the ER to the cytosol resulting in early apoptotic cell death. These mechanistic insights suggest why this class of BH3-only mimetic synergizes in a particular way with Pseudomonas exotoxin-based immunotoxins.


Assuntos
Adenocarcinoma/tratamento farmacológico , Compostos de Bifenilo/administração & dosagem , Exotoxinas/administração & dosagem , Imunotoxinas/administração & dosagem , Nitrofenóis/administração & dosagem , Sulfonamidas/administração & dosagem , Neoplasias do Colo do Útero/tratamento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/patologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptose/efeitos dos fármacos , Citosol , Retículo Endoplasmático/efeitos dos fármacos , Exotoxinas/genética , Feminino , Humanos , Imunotoxinas/genética , Piperazinas/administração & dosagem , Biossíntese de Proteínas , Pseudomonas/metabolismo , Neoplasias do Colo do Útero/diagnóstico , Neoplasias do Colo do Útero/patologia
15.
J Immunother ; 37(1): 8-15, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24316551

RESUMO

SS1P is an antimesothelin recombinant immunotoxin (RIT). Pancreatic ductal adenocarcinoma (PDAC) cell lines are resistant to SS1P, despite high mesothelin expression. The aim of this study is to examine whether combining SS1P and BH3-mimetic ABT-737 induces cell death in a panel of PDAC cell lines. ABT-737 binds and neutralizes several antiapoptotic BCL2 family proteins, but has a low affinity for the short-lived MCL1 and BCL2A1. SS1P inhibits protein synthesis, which has shown to downregulate MCL1. PDAC cell lines KLM-1, BxPc-3, and Panc 3.014 were resistant to SS1P or ABT-737 alone. Combining both compounds led to a significant increase in cell death. After 48 hours of treatment, cell death was observed in 92% of KLM-1, 55% of BxPc-3, and 23% of Panc 3.014 cells. Panc 3.014 had the highest number of mesothelin-binding sites (92×10(3)), followed by KLM-1 (58×10(3)) and BxPc-3 (3×10(3)). ABT-737 had no effect on SS1P internalization, but enhanced SS1P-induced protein synthesis inhibition significantly in KLM-1, to a lesser extent in BxPc-3, and very little in Panc 3.014. SS1P alone or in combination with ABT-737 downregulated MCL1 in KLM-1 and BxPc-3, but not in Panc 3.014. Similar observations were made for BCL2A1, which had the highest levels in Panc 3.014. Compared with KLM-1, Panc 3.014, and BxPc-3 also had lower proapoptotic BAK and a trend toward higher MCL1. Proapoptotic BAX was similar in KLM-1 and BxPc-3, but lower in Panc 3.014. In conclusion, combining SS1P with ABT-737 overcomes SS1P-resistance in PDAC, although to a variable extent. The efficacy of the combination is mainly associated with the RIT-associated inhibition of protein synthesis and the ability to downregulate MCL1 and BCL2A1, while levels of other key apoptotic proteins may also be important. Our data support the combination of an RIT and a BH3-mimetic, and identify factors that potentially limit the efficacy of such therapeutic approach.


Assuntos
Anticorpos Monoclonais/farmacologia , Compostos de Bifenilo/farmacologia , Resistencia a Medicamentos Antineoplásicos , Proteínas Ligadas por GPI/antagonistas & inibidores , Imunotoxinas/farmacologia , Nitrofenóis/farmacologia , Neoplasias Pancreáticas/imunologia , Sulfonamidas/farmacologia , Anticorpos Monoclonais/imunologia , Antígenos de Neoplasias/imunologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citotoxicidade Imunológica/efeitos dos fármacos , Sinergismo Farmacológico , Proteínas Ligadas por GPI/imunologia , Humanos , Imunotoxinas/imunologia , Mesotelina , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/terapia , Piperazinas/farmacologia , Ligação Proteica/imunologia , Biossíntese de Proteínas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo
16.
Toxins (Basel) ; 5(8): 1486-502, 2013 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-23965432

RESUMO

Immunotoxins are antibody-toxin bifunctional molecules that rely on intracellular toxin action to kill target cells. Target specificity is determined via the binding attributes of the chosen antibody. Mostly, but not exclusively, immunotoxins are purpose-built to kill cancer cells as part of novel treatment approaches. Other applications for immunotoxins include immune regulation and the treatment of viral or parasitic diseases. Here we discuss the utility of protein toxins, of both bacterial and plant origin, joined to antibodies for targeting cancer cells. Finally, while clinical goals are focused on the development of novel cancer treatments, much has been learned about toxin action and intracellular pathways. Thus toxins are considered both medicines for treating human disease and probes of cellular function.


Assuntos
Imunotoxinas/uso terapêutico , Animais , Anticorpos Monoclonais/uso terapêutico , Toxinas Bacterianas/uso terapêutico , Quimioterapia Combinada , Humanos , Neoplasias/terapia , Proteínas Recombinantes de Fusão/uso terapêutico
17.
Leuk Lymphoma ; 52 Suppl 2: 79-81, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21599608

RESUMO

Immunotoxins are antibody-toxin fusion proteins directed to kill cancer cells displaying specific target antigens on their surface. Remarkably, immunotoxins directed to CD22 on hairy cell leukemia have produced complete remissions in approximately 60% of patients enrolled in phase I/II trials. For reasons that are not yet clear, 40% of patients responded less well. In addition, patients with other CD22-positive malignancies have not yet achieved complete remissions. In trying to understand 'resistance' to immunotoxin therapy, a number of challenging issues have been raised. These include insufficient dosing, the production of neutralizing anti-immunotoxin antibodies, poor access to malignant cells, and resistance to toxin killing. In designing immunotoxins, we employ truncated Pseudomonas exotoxin, which enzymatically inactivates protein synthesis and produces cell death in sensitive cells. To begin to address toxin resistance we have explored combination therapy with the BH3-only mimetic, ABT-737. Our results indicate that immunotoxin-ABT combinations often exhibit greater killing activity than either compound alone and in some instances overcome resistance. Expression of high levels of prosurvival Bcl-2 proteins may contribute to toxin resistance.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Leucemia de Células Pilosas/tratamento farmacológico , Compostos de Bifenilo/uso terapêutico , Sinergismo Farmacológico , Humanos , Imunotoxinas/uso terapêutico , Nitrofenóis/uso terapêutico , Piperazinas/uso terapêutico , Lectina 2 Semelhante a Ig de Ligação ao Ácido Siálico/imunologia , Sulfonamidas/uso terapêutico
18.
Mol Cancer Ther ; 9(7): 2007-15, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20587662

RESUMO

Pseudomonas exotoxin (PE)-based immunotoxins (antibody-toxin fusion proteins) have achieved frequent complete remissions in patients with hairy cell leukemia but far fewer objective responses in other cancers. To address possible mechanisms of resistance, we investigated immunotoxin activity in a model system using the colon cancer cell line, DLD1. Despite causing complete inhibition of protein synthesis, there was no evidence that an immunotoxin targeted to the transferrin receptor caused apoptosis in these cells. To address a possible protective role of prosurvival Bcl-2 proteins, the BH3-only mimetic, ABT-737, was tested alone or in combination with immunotoxins. Neither the immunotoxin nor ABT-737 alone activated caspase 3, whereas the combination exhibited substantial activation. In other epithelial cell lines, ABT-737 enhanced the cytotoxicity of PE-related immunotoxins by as much as 20-fold, but did not enhance diphtheria toxin or cycloheximide. Because PE translocates to the cytosol via the endoplasmic reticulum (ER) and the other toxins do not, ABT-737-mediated effects on the ER were investigated. ABT-737 treatment stimulated increased levels of ER stress response factor, ATF4. Because of its activity in the ER, ABT-737 might be particularly well suited for enhancing the activity of immunotoxins that translocate from the ER to the cell cytosol.


Assuntos
Apoptose/efeitos dos fármacos , Compostos de Bifenilo/farmacologia , Imunotoxinas/farmacologia , Nitrofenóis/farmacologia , Sulfonamidas/farmacologia , Fator 4 Ativador da Transcrição/metabolismo , Western Blotting , Caspases/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citosol/efeitos dos fármacos , Citosol/metabolismo , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Sinergismo Farmacológico , Retículo Endoplasmático/metabolismo , Exotoxinas/metabolismo , Exotoxinas/farmacologia , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , Piperazinas/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Pseudomonas/metabolismo
19.
Cancer Immunol Immunother ; 59(5): 737-46, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20091030

RESUMO

Immunotoxins are antibody-toxin fusion proteins under development as cancer therapeutics. In early clinical trials, immunotoxins constructed with domains II and III of Pseudomonas exotoxin (termed PE38), have produced a high rate of complete remissions in Hairy Cell Leukemia and objective responses in other malignancies. Cholera exotoxin (also known as cholix toxin) has a very similar three-dimensional structure to Pseudomonas exotoxin (PE) and when domains II and III of each are compared at the primary sequence level, they are 36% identical and 50% similar. Here we report on the construction and activity of an immunotoxin made with domains II and III of cholera exotoxin (here termed CET40). In cell viability assays, the CET40 immunotoxin was equipotent to tenfold less active compared to a PE-based immunotoxin made with the same single-chain Fv. A major limitation of toxin-based immunotoxins is the development of neutralizing antibodies to the toxin portion of the immunotoxin. Because of structure and sequence similarities, we evaluated a CET40 immunotoxin for the presence of PE-related epitopes. In western blots, three-of-three anti-PE antibody preparations failed to react with the CET40 immunotoxin. More importantly, in neutralization studies neither these antibodies nor those from patients with neutralizing titers to PE38, neutralized the CET40-immunotoxin. We propose that the use of modular components such as antibody Fvs and toxin domains will allow a greater flexibility in how these agents are designed and deployed including the sequential administration of a second immunotoxin after patients have developed neutralizing antibodies to the first.


Assuntos
Toxina da Cólera/imunologia , Imunotoxinas/imunologia , Sequência de Aminoácidos , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Sequência de Bases , Western Blotting , Toxina da Cólera/genética , Reações Cruzadas , Exotoxinas/imunologia , Humanos , Imunotoxinas/genética , Dados de Sequência Molecular , Neoplasias/sangue , Neoplasias/imunologia , Pseudomonas/imunologia , Receptores da Transferrina/imunologia , Proteínas Recombinantes de Fusão/genética , Anticorpos de Cadeia Única/imunologia
20.
J Biol Chem ; 284(49): 34390-9, 2009 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-19805544

RESUMO

Endophilins participate in membrane scission events that occur during endocytosis and intracellular organelle biogenesis through the combined activity of an N-terminal BAR domain that interacts with membranes and a C-terminal SH3 domain that mediates protein binding. Endophilin B1 (Endo B1) was identified to bind Bax, a Bcl-2 family member that promotes apoptosis, through yeast two-hybrid protein screens. Although Endo B1 does not bind Bax in healthy cells, during apoptosis, Endo B1 interacts transiently with Bax and promotes cytochrome c release from mitochondria. To explore the molecular mechanism of action of Endo B1, we have analyzed its interaction with Bax in cell-free systems. Purified recombinant Endo B1 in solution displays a Stokes radius indicating a tetrameric quarternary structure. However, when incubated with purified Bax, it assembles into oligomers more than 4-fold greater in molecular weight. Although Endo B1 oligomerization is induced by Bax, Bax does not stably associate with the high molecular weight Endo B1 complex. Endo B1 oligomerization requires its C-terminal Src homology 3 domain and is not induced by Bcl-xL. Endo B1 combined with Bax reduces the size and changes the morphology of giant unilamellar vesicles by inducing massive vesiculation of liposomes. This activity of purified Bax protein to induce cell-free assembly of Endo B1 may reflect its activity in cells that regulates apoptosis and/or mitochondrial fusion.


Assuntos
Aciltransferases/química , Lipídeos de Membrana/química , Proteína X Associada a bcl-2/química , Apoptose , Catálise , Sistema Livre de Células , DNA Complementar/metabolismo , Endocitose , Humanos , Lipossomos/química , Microscopia de Fluorescência/métodos , Modelos Estatísticos , Estrutura Quaternária de Proteína , Estrutura Terciária de Proteína , Proteína bcl-X/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...