Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Toxins (Basel) ; 14(8)2022 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-36006226

RESUMEN

Targeted toxins (TT) for cancer treatment are a class of hybrid biologic comprised of a targeting domain coupled chemically or genetically to a proteinaceous toxin payload. The targeting domain of the TT recognises and binds to a defined target molecule on the cancer cell surface, thereby delivering the toxin that is then required to internalise to an appropriate intracellular compartment in order to kill the target cancer cell. Toxins from several different sources have been investigated over the years, and the two TTs that have so far been licensed for clinical use in humans; both utilise bacterial toxins. Relatively few clinical studies have, however, been undertaken with TTs that utilise single-chain type I ribosome inactivating proteins (RIPs). This paper reviews the clinical experience that has so far been obtained for a range of TTs based on five different type I RIPs and concludes that the majority studied in early phase trials show significant clinical activity that justifies further clinical investigation. A range of practical issues relating to the further clinical development of TT's are also covered briefly together with some suggested solutions to outstanding problems.


Asunto(s)
Inmunotoxinas , Proteínas de Plantas/uso terapéutico , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Toxinas Biológicas , Humanos , Inmunotoxinas/uso terapéutico , Neoplasias/tratamiento farmacológico , Proteínas de Plantas/metabolismo
2.
Mol Biol Rep ; 48(4): 3827-3840, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33895972

RESUMEN

Ribosome inactivating proteins (RIPs) as family of anti-cancer drugs recently received much attention due to their interesting anti-cancer mechanism. In spite of small drugs, RIPs use the large-size effect (LSE) to prevent the efflux process governed by drug resistance transporters (DRTs) which prevents inside of the cells against drug transfection. There are many clinical translation obstacles that severely restrict their applications especially their delivery approach to the tumor cells. As the main goal of this review, we will focus on trichosanthin (TCS) and gelonin (Gel) and other types, especially scorpion venom-derived RIPs to clarify that they are struggling with what types of bio-barriers and these challenges could be solved in cancer therapy science. Then, we will try to highlight recent state-of-the-arts in delivery of RIPs for cancer therapy.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Proteínas Inactivadoras de Ribosomas Tipo 1/toxicidad , Tricosantina/toxicidad , Animales , Antineoplásicos Fitogénicos/toxicidad , Humanos , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Venenos de Escorpión/uso terapéutico , Venenos de Escorpión/toxicidad , Bloqueadores de los Canales de Sodio/uso terapéutico , Bloqueadores de los Canales de Sodio/toxicidad , Tricosantina/uso terapéutico
3.
Toxins (Basel) ; 12(9)2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32957454

RESUMEN

The deadly pandemic named COVID-19, caused by a new coronavirus (SARS-CoV-2), emerged in 2019 and is still spreading globally at a dangerous pace. As of today, there are no proven vaccines, therapies, or even strategies to fight off this virus. Here, we describe the in silico docking results of a novel broad range anti-infective fusion protein RTAM-PAP1 against the various key proteins of SARS-CoV-2 using the latest protein-ligand docking software. RTAM-PAP1 was compared against the SARS-CoV-2 B38 antibody, ricin A chain, a pokeweed antiviral protein from leaves, and the lectin griffithsin using the special CoDockPP COVID-19 version. These experiments revealed novel binding mechanisms of RTAM-PAP1 with a high affinity to numerous SARS-CoV-2 key proteins. RTAM-PAP1 was further characterized in a preliminary toxicity study in mice and was found to be a potential therapeutic candidate. These findings might lead to the discovery of novel SARS-CoV-2 targets and therapeutic protein structures with outstanding functions.


Asunto(s)
Antivirales/química , Antivirales/uso terapéutico , Betacoronavirus/efectos de los fármacos , Infecciones por Coronavirus/tratamiento farmacológico , Neumonía Viral/tratamiento farmacológico , Unión Proteica/efectos de los fármacos , Proteínas Inactivadoras de Ribosomas Tipo 1/química , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Ricina/uso terapéutico , Animales , COVID-19 , Simulación por Computador , Humanos , Ratones , Modelos Animales , Pandemias , Phytolacca americana/química , Hojas de la Planta/química , Proteínas Inactivadoras de Ribosomas Tipo 1/genética , SARS-CoV-2
4.
J Am Chem Soc ; 140(23): 7282-7291, 2018 06 13.
Artículo en Inglés | MEDLINE | ID: mdl-29809001

RESUMEN

The intracellular delivery of biofunctional enzymes or therapeutic proteins through systemic administration is of great importance in therapeutic intervention of various diseases. However, current strategies face substantial challenges owing to various biological barriers, including susceptibility to protein degradation and denaturation, poor cellular uptake, and low transduction efficiency into the cytosol. Here, we developed a biomimetic nanoparticle platform for systemic and intracellular delivery of proteins. Through a biocompatible strategy, guest proteins are caged in the matrix of metal-organic frameworks (MOFs) with high efficiency (up to ∼94%) and high loading content up to ∼50 times those achieved by surface conjunction, and the nanoparticles were further decorated with the extracellular vesicle (EV) membrane with an efficiency as high as ∼97%. In vitro and in vivo study manifests that the EV-like nanoparticles can not only protect proteins against protease digestion and evade the immune system clearance but also selectively target homotypic tumor sites and promote tumor cell uptake and autonomous release of the guest protein after internalization. Assisted by biomimetic nanoparticles, intracellular delivery of the bioactive therapeutic protein gelonin significantly inhibits the tumor growth in vivo and increased 14-fold the therapeutic efficacy. Together, our work not only proposes a new concept to construct a biomimetic nanoplatform but also provides a new solution for systemic and intracellular delivery of protein.


Asunto(s)
Portadores de Fármacos/química , Vesículas Extracelulares/química , Estructuras Metalorgánicas/química , Nanopartículas/química , Proteínas Inactivadoras de Ribosomas Tipo 1/química , Animales , Antineoplásicos/química , Antineoplásicos/uso terapéutico , Materiales Biomiméticos/química , Materiales Biomiméticos/metabolismo , Materiales Biomiméticos/uso terapéutico , Materiales Biomiméticos/toxicidad , Línea Celular Tumoral , Membrana Celular/química , Membrana Celular/metabolismo , Portadores de Fármacos/metabolismo , Portadores de Fármacos/uso terapéutico , Portadores de Fármacos/toxicidad , Endocitosis/fisiología , Vesículas Extracelulares/metabolismo , Humanos , Estructuras Metalorgánicas/metabolismo , Estructuras Metalorgánicas/uso terapéutico , Estructuras Metalorgánicas/toxicidad , Ratones , Nanopartículas/metabolismo , Nanopartículas/uso terapéutico , Nanopartículas/toxicidad , Neoplasias/tratamiento farmacológico , Tamaño de la Partícula , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
5.
ACS Chem Biol ; 12(11): 2891-2897, 2017 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-29019389

RESUMEN

With the high efficacy of protein-based therapeutics and plenty of intracellular drug targets, cytosolic protein delivery in a cell-specific manner has attracted considerable attention in the field of precision medicine. Herein, we present an intracellular protein delivery system based on a target-specific repebody and the translocation domain of Pseudomonas aeruginosa exotoxin A. The delivery platform was constructed by genetically fusing an EGFR-specific repebody as a targeting moiety to the translocation domain, while a protein cargo was fused to the C-terminal end of the delivery platform. The delivery platform was revealed to efficiently translocate a protein cargo to the cytosol in a target-specific manner. We demonstrate the utility and potential of the delivery platform by showing a remarkable tumor regression with negligible toxicity in a xenograft mice model when gelonin was used as the cytotoxic protein cargo. The present platform can find wide applications to the cell-selective cytosolic delivery of diverse proteins in many areas.


Asunto(s)
ADP Ribosa Transferasas/química , Antineoplásicos Fitogénicos/administración & dosificación , Toxinas Bacterianas/química , Portadores de Fármacos/química , Exotoxinas/química , Neoplasias/tratamiento farmacológico , Proteínas Inactivadoras de Ribosomas Tipo 1/administración & dosificación , Factores de Virulencia/química , Animales , Antineoplásicos Fitogénicos/uso terapéutico , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Modelos Moleculares , Dominios Proteicos , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Exotoxina A de Pseudomonas aeruginosa
6.
PLoS One ; 12(10): e0184496, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29020090

RESUMEN

Antigen-specific T-cells are highly variable, spanning potent antiviral efficacy and damaging auto-reactivity. In virus infections, identifying the most efficacious responses is critical to vaccine design. However, current methods depend on indirect measures or on ex vivo expanded CTL clones. We here describe a novel application of cytotoxic saporin-conjugated tetramers to kill antigen-specific T-cells without significant off-target effects. The relative efficacy of distinct antiviral CD8+ T-cell specificity can be directly assessed via antigen-specific CD8+ T-cell depletion. The utility of these reagents is demonstrated here in identifying the CD8+ T-cell specificity most effective in preventing HIV progression in HIV-infected HLA-B*27-positive immune controllers.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , Multimerización de Proteína , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Antivirales/farmacología , Linfocitos T CD8-positivos/efectos de los fármacos , Endocitosis/efectos de los fármacos , Humanos , Depleción Linfocítica , Saporinas
7.
Int Rev Immunol ; 36(4): 207-219, 2017 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-28282218

RESUMEN

Immunotoxins are a novel class of cancer therapeutics that contains a cytotoxic agent fused to a targeting moiety. Various toxic agents from different sources are used in immunotoxin development, including bacterial, plant and human origin cytotoxic elements. Although bacterial and plant-derived toxins are highly toxic and commonly used in immunotoxins, their immunogenicity for human restricted their application in cancer therapy. Here, we discuss the advantages and limitations of bacterial toxins such as Pseudomonas and Diphtheria toxins, plant toxins such as ricin and gelonin, and some endogenous protein of human origin such as RNases and Granzymes. This article will also review different generations of immunotoxins with special focus on immunotoxins which are under clinical trials or approved for clinical use. Finally, current deimmunization strategies for development of new less-immunogenic recombinant immunotoxins will be discussed. ABBREVIATIONS: mAbs: Monoclonal antibodies; EF2: elongation factor 2; ITs: Immunotoxins; DT: Diphtheria toxin; PE: Pseudomonas exotoxin; dgA: de-glycosylated A-chain of ricin; rGel: recombinant de-glycosylated form of gelonin; NKC: natural killer cells; HTR: human transferrin receptor; EGF: epidermal growth factor; GM-CSF: granulocyte-macrophage colony-stimulating factor; DAB389: truncated Diphtheria toxin; B-CCL: B-cell chronic lymphocytic leukemia; RCC: renal cell carcinoma; GVHD: Graft-versus-host disease; EGFR: epidermal growth factor receptor; AML: acute myeloid leukemia; Fab: fragment antigen-binding; dsFv: disulfide-stabilized fragment variable; scFv: single-chain fragment variable; B-ALL: B-lineage Acute Lymphoblastic Leukemia; Fv: fragment variable; HCL: hairy cell leukemia; IL-2R: Interleukin-2 receptor; CR: complete response; CLL: chronic lymphocytic leukemia; ATL: adult T-cell leukemia; DARPins: designed Ankyrin repeat proteins; pmol: picomolar; HAMA: human-anti mouse antibody.


Asunto(s)
Antineoplásicos/uso terapéutico , Inmunoterapia/métodos , Inmunotoxinas/uso terapéutico , Neoplasias/terapia , Animales , Ensayos Clínicos como Asunto , Desensibilización Inmunológica , Toxina Diftérica/uso terapéutico , Granzimas/uso terapéutico , Humanos , Ratones , Neoplasias/inmunología , Ribonucleasas/uso terapéutico , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Ricina/uso terapéutico
8.
Nucleic Acid Ther ; 26(3): 156-65, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27228412

RESUMEN

Delivery of toxins, such as the ricin A chain, Pseudomonas exotoxin, and gelonin, using antibodies has had some success in inducing specific toxicity in cancer treatments. However, these antibody-toxin conjugates, called immunotoxins, can be bulky, difficult to express, and may induce an immune response upon in vivo administration. We previously reported delivery of a recombinant variant of gelonin (rGel) by the full-length prostate-specific membrane antigen (PSMA) binding aptamer, A9, to potentially circumvent some of these problems. Here, we report a streamlined approach to generating aptamer-rGel conjugates utilizing a chemically synthesized minimized form of the A9 aptamer. Unlike the full-length A9 aptamer, this minimized variant can be chemically synthesized with a 5' terminal thiol. This facilitates the large scale synthesis and generation of aptamer toxin conjugates linked by a reducible disulfide linkage. Using this approach, we generated aptamer-toxin conjugates and evaluated their binding specificity and toxicity. On PSMA(+) LNCaP prostate cancer cells, the A9.min-rGel conjugate demonstrated an IC50 of ∼60 nM. Additionally, we performed a stability analysis of this conjugate in mouse serum where the conjugate displayed a t1/2 of ∼4 h, paving the way for future in vivo experiments.


Asunto(s)
Antígenos de Superficie/administración & dosificación , Glutamato Carboxipeptidasa II/administración & dosificación , Inmunotoxinas/química , Integrina alfa6beta4/administración & dosificación , Neoplasias/tratamiento farmacológico , Proteínas Inactivadoras de Ribosomas Tipo 1/química , Antígenos de Superficie/química , Antígenos de Superficie/genética , Aptámeros de Nucleótidos/administración & dosificación , Aptámeros de Nucleótidos/síntesis química , Aptámeros de Nucleótidos/química , Línea Celular Tumoral , Glutamato Carboxipeptidasa II/química , Glutamato Carboxipeptidasa II/genética , Humanos , Inmunotoxinas/genética , Integrina alfa6beta4/química , Integrina alfa6beta4/genética , Neoplasias/genética , Proteínas Inactivadoras de Ribosomas Tipo 1/genética , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Ricina/uso terapéutico
9.
J Control Release ; 194: 197-210, 2014 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-25204286

RESUMEN

The bottlenecks of current chemotherapy in the treatment of colorectal cancer lie in the ineffectiveness of the existing anti-cancer small molecule drugs as well as the dose-limiting toxicity caused by the nonselective action on normal tissues by such drugs. To address these problems, we introduce a novel therapeutic strategy based on tumor targeting using a non-internalizing anti-carcinoembryonic antigen (CEA) monoclonal antibody (mAb) and intracellular delivery of the extremely potent yet cell-impermeable protein toxin gelonin via the aid of a cell-penetrating peptide (also termed as protein transduction domain; PTD). A chimeric TAT-gelonin fusion protein was genetically engineered, and it displayed remarkably enhanced anti-cancer activity against human colorectal cancer cells, with IC50 values being several orders of magnitude lower than the unmodified gelonin. On the other hand, a chemically synthesized conjugate of heparin and a murine anti-CEA mAb, T84.66 (termed T84.66-Hep) was found able to bind highly specifically to CEA over-expressing LS174T colorectal cancer cells. When mixing together, TAT-gelonin and T84.66-Hep could associate tightly and automatically through an electrostatic interaction between the cationic TAT and anionic heparin. In preliminary in vivo studies using LS174T s.c. xenograft tumor bearing mouse, selective and significantly augmented (58-fold) delivery of TAT-gelonin to the tumor target was observed, when compared with administration of TAT-gelonin alone. More importantly, efficacy studies also revealed that only the TAT-gelonin/T84.66-Hep complex yielded a significant inhibition of tumor growth (46%) without causing gelonin-induced systemic toxicity. Overall, this study suggested a generic strategy to effectively yet safely deliver potent PTD-modified protein toxins to the tumor.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/metabolismo , Antineoplásicos Fitogénicos/administración & dosificación , Antineoplásicos Fitogénicos/metabolismo , Antígeno Carcinoembrionario/metabolismo , Péptidos de Penetración Celular/uso terapéutico , Cartilla de ADN/administración & dosificación , Sistemas de Liberación de Medicamentos , Productos del Gen tat/química , Vectores Genéticos , Heparina/química , Humanos , Masculino , Dosis Máxima Tolerada , Ratones , Ratones Desnudos , Unión Proteica , Proteínas Inactivadoras de Ribosomas Tipo 1/administración & dosificación , Proteínas Inactivadoras de Ribosomas Tipo 1/metabolismo , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Antiviral Res ; 108: 173-80, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24929084

RESUMEN

Chikungunya virus (CHIKV) outbreaks have led to a serious economic burden, as the available treatment strategies can only alleviate disease symptoms, and no effective therapeutics or vaccines are currently available for human use. Here, we report the use of a new cost-effective approach involving production of a recombinant antiviral peptide-fusion protein that is scalable for the treatment of CHIKV infection. A peptide-fusion recombinant protein LATA-PAP1-THAN that was generated by joining Latarcin (LATA) peptide with the N-terminus of the PAP1 antiviral protein, and the Thanatin (THAN) peptide to the C-terminus, was produced in Escherichia coli as inclusion bodies. The antiviral LATA-PAP1-THAN protein showed 89.0% reduction of viral plaque formation compared with PAP1 (46.0%), LATA (67.0%) or THAN (79.3%) peptides alone. The LATA-PAP1-THAN protein reduced the viral RNA load that was 0.89-fold compared with the untreated control cells. We also showed that PAP1 resulted in 0.44-fold reduction, and THAN and LATA resulting in 0.78-fold and 0.73-fold reductions, respectively. The LATA-PAP1-THAN protein inhibited CHIKV replication in the Vero cells at an EC50 of 11.2µg/ml, which is approximately half of the EC50 of PAP1 (23.7µg/ml) and protected the CHIKV-infected mice at the dose of 0.75mg/ml. We concluded that production of antiviral peptide-fusion protein in E. coli as inclusion bodies could accentuate antiviral activities, enhance cellular internalisation, and could reduce product toxicity to host cells and is scalable to epidemic response quantities.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/uso terapéutico , Antivirales/uso terapéutico , Fiebre Chikungunya/prevención & control , Virus Chikungunya/efectos de los fármacos , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Venenos de Araña/uso terapéutico , Animales , Péptidos Catiónicos Antimicrobianos/genética , Péptidos Catiónicos Antimicrobianos/farmacología , Antivirales/farmacología , Fiebre Chikungunya/tratamiento farmacológico , Virus Chikungunya/fisiología , Chlorocebus aethiops , Modelos Animales de Enfermedad , Escherichia coli/genética , Expresión Génica , Ratones Endogámicos ICR , Pruebas de Sensibilidad Microbiana , Proteínas Asociadas a Pancreatitis , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/uso terapéutico , Proteínas Inactivadoras de Ribosomas Tipo 1/genética , Proteínas Inactivadoras de Ribosomas Tipo 1/farmacología , Venenos de Araña/genética , Venenos de Araña/farmacología , Resultado del Tratamiento , Células Vero , Carga Viral , Ensayo de Placa Viral , Replicación Viral/efectos de los fármacos
11.
Angew Chem Int Ed Engl ; 53(11): 2893-8, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24519972

RESUMEN

An efficient and safe method to deliver active proteins into the cytosol of targeted cells is highly desirable to advance protein-based therapeutics. A novel protein delivery platform has been created by combinatorial design of cationic lipid-like materials (termed "lipidoids"), coupled with a reversible chemical protein engineering approach. Using ribonuclease A (RNase A) and saporin as two representative cytotoxic proteins, the combinatorial lipidoids efficiently deliver proteins into cancer cells and inhibit cell proliferation. A study of the structure-function relationship reveals that the electrostatic and hydrophobic interactions between the lipidoids and the protein play a vital role in the formation of protein-lipidoid nanocomplexes and intracellular delivery. A representative lipidoid (EC16-1) protein nanoparticle formulation inhibits cell proliferation in vitro and suppresses tumor growth in a murine breast cancer model.


Asunto(s)
Antineoplásicos/administración & dosificación , Lípidos/química , Nanopartículas/química , Neoplasias/tratamiento farmacológico , Ácido Aconítico/análogos & derivados , Ácido Aconítico/química , Alcanos/química , Aminas/química , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Humanos , Ratones , Ribonucleasa Pancreática/metabolismo , Ribonucleasa Pancreática/uso terapéutico , Ribonucleasa Pancreática/toxicidad , Proteínas Inactivadoras de Ribosomas Tipo 1/metabolismo , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Proteínas Inactivadoras de Ribosomas Tipo 1/toxicidad , Saporinas
13.
Anesthesiology ; 119(5): 1178-85, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24195949

RESUMEN

BACKGROUND: Substance P-saporin (SP-SAP), a chemical conjugate of substance P and a recombinant version of the ribosome-inactivating protein, saporin, when administered intrathecally, acts as a targeted neurotoxin producing selective destruction of superficial neurokinin-1 receptor-bearing cells in the spinal dorsal horn. The goal of this study was to provide proof-of-concept data that a single intrathecal injection of SP-SAP could safely provide effective pain relief in spontaneous bone cancer pain in companion (pet) dogs. METHODS: In a single-blind, controlled study, 70 companion dogs with bone cancer pain were randomized to standard-of-care analgesic therapy alone (control, n=35) or intrathecal SP-SAP (20-60 µg) in addition to standard-of-care analgesic therapy (n=35). Activity, pain scores, and videography data were collected at baseline, 2 weeks postrandomization, and then monthly until death. RESULTS: Although the efficacy results at the 2-week postrandomization point were equivocal, the outcomes evaluated beyond 2 weeks revealed a positive effect of SP-SAP on chronic pain management. Significantly, more dogs in the control group (74%) required unblinding and adjustment in analgesic protocol or euthanasia within 6 weeks of randomization than dogs that were treated with SP-SAP (24%; P<0.001); and overall, dogs in the control group required unblinding significantly sooner than dogs that had been treated with SP-SAP (P<0.01). CONCLUSION: Intrathecal administration of SP-SAP in dogs with bone cancer produces a time-dependent antinociceptive effect with no evidence of development of deafferentation pain syndrome which can be seen with neurolytic therapies.


Asunto(s)
Neoplasias Óseas/complicaciones , Neoplasias Óseas/veterinaria , Enfermedades de los Perros/tratamiento farmacológico , Dolor Intratable/tratamiento farmacológico , Dolor Intratable/veterinaria , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Sustancia P/análogos & derivados , Animales , Conducta Animal , Perros , Método Doble Ciego , Inyecciones Espinales , Cojera Animal/tratamiento farmacológico , Cojera Animal/psicología , Actividad Motora/fisiología , Dimensión del Dolor/efectos de los fármacos , Dolor Intratable/etiología , Estudios Prospectivos , Saporinas , Sustancia P/uso terapéutico , Resultado del Tratamiento
14.
J Control Release ; 172(1): 169-178, 2013 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-23973813

RESUMEN

The ineffectiveness of small molecule drugs against cancer has generated significant interest in more potent macromolecular agents. Gelonin, a plant-derived toxin that inhibits protein translation, has attracted much attention in this regard. Due to its inability to internalize into cells, however, gelonin exerts only limited tumoricidal effect. To overcome this cell membrane barrier, we modified gelonin, via both chemical conjugation and genetic recombination methods, with low molecular weight protamine (LMWP), a cell-penetrating peptide (CPP) which was shown to efficiently ferry various cargoes into cells. Results confirmed that gelonin-LMWP chemical conjugate (cG-L) and recombinant gelonin-LMWP chimera (rG-L) possessed N-glycosidase activity equivalent to that of unmodified recombinant gelonin (rGel); however, unlike rGel, both gelonin-LMWPs were able to internalize into cells. Cytotoxicity studies further demonstrated that cG-L and rG-L exhibited significantly improved tumoricidal effects, with IC50 values being 120-fold lower than that of rGel. Moreover, when tested against a CT26 s.c. xenograft tumor mouse model, significant inhibition of tumor growth was observed with rG-L doses as low as 2 µg/tumor, while no detectable therapeutic effects were seen with rGel at 10-fold higher doses. Overall, this study demonstrated the potential of utilizing CPP-modified gelonin as a highly potent anticancer drug to overcome limitations of current chemotherapeutic agents.


Asunto(s)
Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Proteínas Inactivadoras de Ribosomas Tipo 1/química , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Animales , Antineoplásicos Fitogénicos/metabolismo , Línea Celular Tumoral , Péptidos de Penetración Celular/química , Péptidos de Penetración Celular/genética , Péptidos de Penetración Celular/uso terapéutico , Humanos , Masculino , Ratones , Ratones Desnudos , Neoplasias/patología , Ratas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/uso terapéutico , Proteínas Inactivadoras de Ribosomas Tipo 1/genética , Suregada/química
15.
Eur J Orthop Surg Traumatol ; 23(3): 263-72, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23412299

RESUMEN

We employed 54 rats to devise a model of neuroma-in-continuity and explore the effect of the immunotoxin OX7-saporin on the neuroma. The left common peroneal, tibial or sciatic nerves were crushed by one 10-s application of a micro-artery forceps. At 3 and 6 weeks, the nerve was cut distal to the site of nerve crush, and retrograde fluorescent labeling was done. Pressure microinjection of 2 µl of natural saline or 2 µl of the immunotoxin conjugate OX7-saporin was done at the nerve stump 2 days later. Sacrifice was done after 3 weeks. In all control and saline-injection nerve specimens, gross observation and histology showed a neuroma-in-continuity. In 19 of the 24 OX7-saporin nerve specimens, gross observation showed a narrowed area at the site of nerve crush. Histology showed inhibition of neuroma-in-continuity formation. Fluorescent microscopy showed ablation of the labeled neurons in the dorsal root ganglia corresponding to the OX7-saporin subgroups.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Inmunoconjugados/uso terapéutico , Inmunotoxinas/uso terapéutico , Traumatismos de los Nervios Periféricos/tratamiento farmacológico , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Animales , Anticuerpos Monoclonales/administración & dosificación , Modelos Animales de Enfermedad , Ganglios Espinales/efectos de los fármacos , Inmunoconjugados/administración & dosificación , Inmunotoxinas/administración & dosificación , Masculino , Microinyecciones/métodos , Microscopía Fluorescente , Compresión Nerviosa , Traumatismos de los Nervios Periféricos/patología , Nervio Peroneo/lesiones , Nervio Peroneo/patología , Ratas , Proteínas Inactivadoras de Ribosomas Tipo 1/administración & dosificación , Saporinas , Nervio Ciático/lesiones , Nervio Ciático/patología , Nervio Tibial/lesiones , Nervio Tibial/patología
16.
Haematologica ; 98(2): 217-21, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22875630

RESUMEN

We conducted a phase 1 study of an anti-CD33 immunotoxin, humanized monoclonal antibody M195 conjugated to recombinant gelonin (HUM-195/rGEL), in patients with relapsed, refractory myeloid leukemias. Twenty-eight patients received the construct intravenously at four dose levels (12, 18, 28 and 40 mg/m(2) per course) in a "3+3" study design. The dose-limiting toxicity was infusion-related allergic reaction including hypoxia and hypotension. The 28 mg/m(2) total dose was considered the maximally tolerated dose. Four patients developed a reduction in peripheral blood blasts of at least 50%. Three patients treated with the 10, 12 and 28 mg/m(2) doses showed a 38-50% reduction in bone marrow blasts. There was normalization of platelets in one patient treated with 40 mg/m(2). Pharmacokinetic analysis demonstrated that the highest blood levels achieved were 200-300 ng/mL which cleared with a half-life of ∼20 hours. Antigenicity was low with one patient at the 12 mg/m(2) dose and one patient at the 18 mg/m(2) dose (2/23, <10%) developing antibodies to the recombinant gelonin component after 28 days. We concluded that HUM-195/rGel can be safely administered in a multi-dose cycle to patients with advanced myeloid malignancies and warrants further investigation.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Inmunotoxinas/uso terapéutico , Leucemia Mieloide/tratamiento farmacológico , Leucemia Mieloide/patología , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Adulto , Anciano , Anciano de 80 o más Años , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Femenino , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Inmunotoxinas/inmunología , Inmunotoxinas/farmacología , Leucemia Mieloide/inmunología , Masculino , Persona de Mediana Edad , Proteínas Inactivadoras de Ribosomas Tipo 1/inmunología , Proteínas Inactivadoras de Ribosomas Tipo 1/farmacología , Resultado del Tratamiento
17.
Leuk Res ; 37(1): 83-8, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23040543

RESUMEN

Targeted therapies, such as those using imatinib and rituximab, have revolutionized the treatment of Philadelphia chromosome-positive and CD20-positive acute lymphoblastic leukemia (ALL) respectively, yet these therapies are effective in only a subset of patients and remission is generally not durable. The next generation of targeted therapies includes the use of antibodies conjugated to potent cytotoxic agents and are classified as antibody drug conjugates (ADC). For B-lineage ALL, CD22 is an ideal target for ADC therapy because it is expressed on the majority of B-lineage ALL cells and because antibody binding mediates receptor internalization. HB22.7-SAP is a conjugate of our anti-CD22 monoclonal antibody (mAb), HB22.7, and the ribosome inhibiting protein, saporin (SAP). In vitro, HB22.7-SAP effectively bound to CD22 on the surface of pre-B ALL cell lines and exhibited potent and specific cytotoxicity. In a NOD/SCID xenograft mouse model of pre-B ALL, when compared to the vehicle-treated control, HB22.7-SAP increased the median survival time from 20 days to over 50 days without significant toxicity.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Inmunotoxinas/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Animales , Femenino , Humanos , Ratones , Ratones SCID , Trasplante de Neoplasias , Saporinas , Trasplante Heterólogo
18.
J Control Release ; 164(1): 74-86, 2012 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-23063550

RESUMEN

Type I ribosome inactivating proteins such as saporin from the plant Saponaria officinalis L. are widely used as toxin moieties of targeted anti-tumor toxins. For exerting cytotoxicity the toxin moieties have to be released into the cytosol of tumor cells. However the cytosolic transfer of toxin molecules into the cytosol is mostly an inefficient process. In this report we demonstrate that certain saponins, which are also biosynthesized by Saponaria officinalis L., specifically mediate the release of saporin out of the intracellular compartments into the cytosol without affecting the integrity of the plasma membrane. The relevant cellular compartments were identified as late endosomes and lysosomes. Further studies revealed that endosomal acidification is a prerequisite for the saponin-mediated release of saporin. Binding analysis demonstrated an association of the saponins with saporin in a pH-dependent manner. The applicability of the saponin-mediated effect was demonstrated in vivo in a syngeneic tumor model using a saporin-based targeted anti-tumor toxin in combination with characterized saponins.


Asunto(s)
Antineoplásicos Fitogénicos/administración & dosificación , Citosol/metabolismo , Portadores de Fármacos/química , Proteínas Inactivadoras de Ribosomas Tipo 1/administración & dosificación , Saponinas/química , Animales , Antineoplásicos Fitogénicos/farmacocinética , Antineoplásicos Fitogénicos/uso terapéutico , Antineoplásicos Fitogénicos/toxicidad , Línea Celular , Supervivencia Celular/efectos de los fármacos , Rastreo Celular , Portadores de Fármacos/farmacocinética , Portadores de Fármacos/toxicidad , Endocitosis , Factor de Crecimiento Epidérmico/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos BALB C , Estructura Molecular , Unión Proteica , Transporte de Proteínas , Proteínas Inactivadoras de Ribosomas Tipo 1/farmacocinética , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Proteínas Inactivadoras de Ribosomas Tipo 1/toxicidad , Saponinas/farmacocinética , Saponinas/toxicidad , Saporinas , Análisis de la Célula Individual , Resonancia por Plasmón de Superficie , Distribución Tisular , Pruebas de Toxicidad Aguda , Ensayos Antitumor por Modelo de Xenoinjerto
19.
J Immunotoxicol ; 9(4): 368-73, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22506556

RESUMEN

It has been shown that naturally occurring regulatory T-cells (CD4(+)CD25(+) Foxp3(+) T-cells) have critical roles in tumor invasion and down-regulation of immune response against established tumors. High expression of CD25 (IL-2Rα) by regulatory T (T(reg)) cells may cause an inefficient response when using IL-2-based cancer vaccines. It seems that selective elimination of T(reg) cells before treatment of tumor-bearing T-cells can strongly increase the efficacy of a vaccine. The aim of this study was to set up an efficient cost-effective protocol to eliminate CD4(+)CD25(+) T-cells-using the immunotoxin anti-tac-SAP. Peripheral blood mononuclear cells (PBMC) taken from colon cancer patients were treated with different concentrations (i.e., 0-100 µg/dl) of the immunotoxin. Flow cytometric analyses were then preformed to analyze expression of CD4, CD25, CD3, CD8, and CD45 surface markers; semi-quantitative fluorescent-PCR was used for the detection of Foxp3 expression before and after anti-tac-SAP treatment. The results indicated that anti-tac-SAP effectively eliminated CD4(+)CD25(+) T(reg) cells and that 25 µg/dl was the optimal concentration of anti-tac-SAP for selective depletion of these cells. These outcomes were verified by analyses of Foxp3 expression. The results also indicated that this immunotoxin had no non-specific effects on other T-cells, including CD4(+)CD25(-) and CD8(+)CD45(+) T-cells. Building on the work here, ongoing/future studies with the anti-tac-SAP will focus on functional assessments of the remaining (i.e., non-eliminated) T-cells (i.e., CD8, CD4; using proliferation and peptide sensitization assays) to ascertain if the immunotoxin inadvertently alters the functions of these cells-an untoward outcome.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Neoplasias del Colon/terapia , Inmunotoxinas/uso terapéutico , Subunidad alfa del Receptor de Interleucina-2/inmunología , Depleción Linfocítica/métodos , Proteínas Recombinantes de Fusión/uso terapéutico , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Saponinas/uso terapéutico , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , ADP Ribosa Transferasas/genética , Anticuerpos Monoclonales/genética , Toxinas Bacterianas/genética , Antígenos CD4/inmunología , Células Cultivadas , Exotoxinas/genética , Citometría de Flujo , Factores de Transcripción Forkhead/inmunología , Humanos , Inmunotoxinas/genética , Proteínas Recombinantes de Fusión/genética , Saponinas/genética , Subgrupos de Linfocitos T/efectos de los fármacos , Linfocitos T Reguladores/efectos de los fármacos , Factores de Virulencia/genética , Exotoxina A de Pseudomonas aeruginosa
20.
Leukemia ; 26(8): 1786-96, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22373785

RESUMEN

Acute lymphoblastic leukemia (ALL) is the most common malignancy affecting children and a major cause of mortality from hematopoietic malignancies in adults. A substantial number of patients become drug resistant during chemotherapy, necessitating the development of alternative modes of treatment. rGel (recombinant Gelonin)/BlyS (B-lymphocyte stimulator) is a toxin-cytokine fusion protein used for selective killing of malignant B-cells expressing receptors for B-cell-activating factor (BAFF/BLyS) by receptor-targeted delivery of the toxin, Gelonin. Here, we demonstrate that rGel/BLyS binds to ALL cells expressing BAFF receptor (BAFF-R) and upon internalization, it induces apoptosis of these cells and causes downregulation of survival genes even in the presence of stromal protection. Using an immunodeficient transplant model for human ALL, we show that rGel/BLyS prolongs survival of both Philadelphia chromosome-positive and negative ALL-bearing mice. Furthermore, we used AMD3100, a CXCR4 antagonist, to mobilize the leukemic cells protected in the bone marrow (BM) microenvironment and the combination with rGel/BLyS resulted in a significant reduction of the tumor load in the BM and complete eradication of ALL cells from the circulation. Thus, a combination treatment with the B-cell-specific fusion toxin rGel/BLyS and the mobilizing agent AMD3100 could be an effective alternative approach to chemotherapy for the treatment of primary and relapsed ALL.


Asunto(s)
Antineoplásicos/uso terapéutico , Factor Activador de Células B/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/tratamiento farmacológico , Proteínas Recombinantes de Fusión/uso terapéutico , Proteínas Inactivadoras de Ribosomas Tipo 1/uso terapéutico , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/metabolismo , Apoptosis/efectos de los fármacos , Factor Activador de Células B/administración & dosificación , Factor Activador de Células B/genética , Factor Activador de Células B/metabolismo , Bencilaminas , Médula Ósea/patología , Línea Celular Tumoral , Ciclamas , Sinergismo Farmacológico , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Compuestos Heterocíclicos/farmacología , Humanos , Ratones , Ratones Desnudos , Ratones SCID , FN-kappa B/antagonistas & inhibidores , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Inactivadoras de Ribosomas Tipo 1/administración & dosificación , Proteínas Inactivadoras de Ribosomas Tipo 1/genética , Proteínas Inactivadoras de Ribosomas Tipo 1/metabolismo , Toxinas Biológicas/administración & dosificación , Toxinas Biológicas/metabolismo , Toxinas Biológicas/uso terapéutico , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA