Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Cell Rep ; 42(1): 111955, 2023 01 31.
Artículo en Inglés | MEDLINE | ID: mdl-36640333

RESUMEN

Delivery of cancer therapeutics to non-specific sites decreases treatment efficacy while increasing toxicity. In ovarian cancer, overexpression of the cell surface marker HER2, which several therapeutics target, relates to poor prognosis. We recently reported the assembly of biocompatible bacterial spore-like particles, termed "SSHELs." Here, we modify SSHELs with an affibody directed against HER2 and load them with the chemotherapeutic agent doxorubicin. Drug-loaded SSHELs reduce tumor growth and increase survival with lower toxicity in a mouse tumor xenograft model compared with free drug and with liposomal doxorubicin by preferentially accumulating in the tumor mass. Target cells actively internalize and then traffic bound SSHELs to acidic compartments, whereupon the cargo is released to the cytosol in a pH-dependent manner. We propose that SSHELs represent a versatile strategy for targeted drug delivery, especially in cancer settings.


Asunto(s)
Neoplasias , Esporas Bacterianas , Ratones , Humanos , Animales , Esporas Bacterianas/metabolismo , Sistemas de Liberación de Medicamentos , Membrana Celular/metabolismo , Neoplasias/metabolismo , Proteínas Bacterianas/metabolismo , Bacillus subtilis/metabolismo
2.
Front Immunol ; 13: 954992, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36341428

RESUMEN

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.


Asunto(s)
Antineoplásicos Inmunológicos , Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Microambiente Tumoral , Neoplasias Pancreáticas/patología , Carcinoma Ductal Pancreático/patología , Inmunoterapia , Antineoplásicos Inmunológicos/uso terapéutico
3.
Biomolecules ; 10(9)2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32957689

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Inmunotoxinas/inmunología , Neoplasias/inmunología , Receptores de Superficie Celular/inmunología , Toxinas Biológicas/inmunología , Animales , Anticuerpos Monoclonales/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/inmunología , Humanos , Inmunotoxinas/metabolismo , Inmunotoxinas/uso terapéutico , Ligandos , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Unión Proteica , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/metabolismo , Toxinas Biológicas/metabolismo
4.
Blood ; 131(21): 2331-2334, 2018 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-29487070

RESUMEN

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.


Asunto(s)
Antineoplásicos/uso terapéutico , Toxinas Bacterianas/uso terapéutico , Exotoxinas/uso terapéutico , Leucemia de Células Pilosas/tratamiento farmacológico , Leucemia de Células Pilosas/patología , Neoplasia Residual/tratamiento farmacológico , Adulto , Anciano , Antineoplásicos/farmacología , Toxinas Bacterianas/farmacología , Ensayos Clínicos Fase I como Asunto , Monitoreo de Drogas , Exotoxinas/farmacología , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Resultado del Tratamiento
5.
Mol Pain ; 13: 1744806917727657, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28814145

RESUMEN

Abstract: Cell deletion approaches to pain directed at either the primary nociceptive afferents or second-order neurons are highly effective analgesic manipulations. Second-order spinal neurons expressing the neurokinin 1 (NK1) receptor are required for the perception of many types of pain. To delete NK1+ neurons for the purpose of pain control, we generated a toxin­peptide conjugate using DTNB-derivatized (Cys0) substance P (SP) and a N-terminally truncated Pseudomonas exotoxin (PE35) that retains the endosome-release and ADP-ribosylation enzymatic domains but with only one free sulfhydryl side chain for conjugation. This allowed generation of a one-to-one product linked by a disulfide bond (SP-PE35). In vitro, Chinese hamster ovary cells stably transfected with the NK1 receptor exhibited specific cytotoxicity when exposed to SP-PE35 (IC50 = 5 × 10−11 M), whereas the conjugate was nontoxic to NK2 and NK3 receptor-bearing cell lines. In vivo studies showed that, after infusion into the spinal subarachnoid space, the toxin was extremely effective in deleting NK1 receptor-expressing cells from the dorsal horn of the spinal cord. The specific cell deletion robustly attenuated thermal and mechanical pain sensations and inflammatory hyperalgesia but did not affect motoric capabilities. NK1 receptor cell deletion and antinociception occurred without obvious lesion of non­receptor-expressing cells or apparent reorganization of primary afferent innervation. These data demonstrate the extraordinary selectivity and broad-spectrum antinociceptive efficacy of this ligand-directed protein therapeutic acting via receptor-mediated endocytosis. The loss of multiple pain modalities including heat and mechanical pinch, transduced by different populations of primary afferents, shows that spinal NK1 receptor-expressing neurons are critical points of convergence in the nociceptive transmission circuit. We further suggest that therapeutic end points can be effectively and safely achieved when SP-PE35 is locally infused, thereby producing a regionally defined analgesia.


Asunto(s)
Exotoxinas/farmacología , Neuronas/metabolismo , Pseudomonas/metabolismo , Receptores de Neuroquinina-1/metabolismo , Animales , Axones/metabolismo , Células CHO , Cricetulus , Hiperalgesia/metabolismo , Dolor/metabolismo , Manejo del Dolor , Sustancia P/metabolismo
6.
PLoS One ; 11(8): e0161415, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27556570

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales , Descubrimiento de Drogas , Ensayos de Selección de Medicamentos Antitumorales , Proteínas Recombinantes de Fusión/farmacología , Toxinas Biológicas , Animales , Anticuerpos Monoclonales/genética , Antineoplásicos/farmacología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Descubrimiento de Drogas/métodos , Exotoxinas/genética , Femenino , Humanos , Inmunotoxinas/genética , Inmunotoxinas/farmacología , Ratones , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Proteínas Recombinantes de Fusión/genética , Bibliotecas de Moléculas Pequeñas , Toxinas Biológicas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Cancer Res ; 76(6): 1560-8, 2016 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-26719540

RESUMEN

Recombinant immunotoxins (RIT) have been highly successful in cancer therapy due, in part, to the high cancer-specific expression of cell surface antigens such as mesothelin, which is overexpressed in mesothelioma, ovarian, lung, breast, and pancreatic cancers, but is limited in normal cells. RG7787 is a clinically optimized RIT consisting of a humanized anti-mesothelin Fab fused to domain III of Pseudomonas exotoxin A, in which immunogenic B-cell epitopes are silenced. To enhance the therapeutic efficacy of RITs, we conducted a kinome RNAi sensitization screen, which identified discoidin domain receptor 1 (DDR1), a collagen-activated tyrosine kinase, as a potential target. The collagen/DDR1 axis is implicated in tumor-stromal interactions and potentially affects tumor response to therapy. Therefore, we investigated the effects of DDR1 on RIT. Knockdown of DDR1 by siRNA or treatment with inhibitor, 7rh, greatly enhanced the cytotoxic activity of RG7787 in several cancer cell lines. Investigation into the mechanism of action showed DDR1 silencing was associated with decreased expression of several ribosomal proteins and enhanced inhibition of protein synthesis. Conversely, induction of DDR1 expression or collagen-stimulated DDR1 activity protected cancer cells from RG7787 killing. Moreover, the combination of RG7787 and DDR1 inhibitor caused greater shrinkage of tumor xenografts than either agent alone. These data demonstrate that DDR1 is a key modulator of RIT activity and represents a novel therapeutic strategy to improve targeting of mesothelin-expressing cancers.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Ligadas a GPI/metabolismo , Inmunotoxinas/farmacología , Proteínas Tirosina Quinasas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , ADP Ribosa Transferasas/farmacología , Animales , Toxinas Bacterianas/farmacología , Línea Celular Tumoral , Receptor con Dominio Discoidina 1 , Exotoxinas/farmacología , Silenciador del Gen/efectos de los fármacos , Humanos , Inmunoconjugados/farmacología , Mesotelina , Ratones , Ratones Desnudos , Factores de Virulencia/farmacología , Exotoxina A de Pseudomonas aeruginosa
8.
Proc Natl Acad Sci U S A ; 112(10): E1135-42, 2015 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-25713356

RESUMEN

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.


Asunto(s)
Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Inmunotoxinas/farmacología , Interferencia de ARN , Animales , Humanos
9.
Blood ; 124(3): 310-2, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-25035145

RESUMEN

In this issue of Blood, Frankel et al describe a novel treatment of blastic plasmacytoid dendritic cell neoplasm (BPDCN) using an engineered version of diphtheria toxin that is targeted to malignant cells via a fusion with interleukin (IL)3 (see panel A).


Asunto(s)
Células Dendríticas/patología , Neoplasias Hematológicas/patología , Neoplasias Hematológicas/terapia , Receptores de Interleucina-3/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/uso terapéutico , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/terapia , Humanos , Masculino
10.
J Immunol ; 193(1): 48-55, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24890727

RESUMEN

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


Asunto(s)
ADP Ribosa Transferasas/farmacología , Formación de Anticuerpos/efectos de los fármacos , Toxinas Bacterianas/farmacología , Exotoxinas/farmacología , Hemocianinas/farmacología , Inmunotoxinas/farmacología , Piperidinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Factores de Virulencia/farmacología , Animales , Linfocitos B/inmunología , Femenino , Centro Germinal/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Quinasas Janus/antagonistas & inhibidores , Quinasas Janus/inmunología , Ratones , Ratones Endogámicos BALB C , Exotoxina A de Pseudomonas aeruginosa
11.
Mol Cancer Ther ; 13(6): 1655-63, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24739394

RESUMEN

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.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Compuestos de Bifenilo/administración & dosificación , Exotoxinas/administración & dosificación , Inmunotoxinas/administración & dosificación , Nitrofenoles/administración & dosificación , Sulfonamidas/administración & dosificación , Neoplasias del Cuello Uterino/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptosis/efectos de los fármacos , Citosol , Retículo Endoplásmico/efectos de los fármacos , Exotoxinas/genética , Femenino , Humanos , Inmunotoxinas/genética , Piperazinas/administración & dosificación , Biosíntesis de Proteínas , Pseudomonas/metabolismo , Neoplasias del Cuello Uterino/diagnóstico , Neoplasias del Cuello Uterino/patología
12.
Blood ; 123(16): 2470-7, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24578503

RESUMEN

Unconjugated monoclonal antibodies that target hematopoietic differentiation antigens have been developed to treat hematologic malignancies. Although some of these have activity against chronic lymphocytic leukemia and hairy cell leukemia, in general, monoclonal antibodies have limited efficacy as single agents in the treatment of leukemia. To increase their potency, the binding domains of monoclonal antibodies can be attached to protein toxins. Such compounds, termed immunotoxins, are delivered to the interior of leukemia cells based on antibody specificity for cell surface target antigens. Recombinant immunotoxins have been shown to be highly cytotoxic to leukemic blasts in vitro, in xenograft model systems, and in early-phase clinical trials in humans. These agents will likely play an increasing role in the treatment of leukemia.


Asunto(s)
Inmunotoxinas/uso terapéutico , Leucemia/terapia , Animales , Antígenos CD19/metabolismo , Ensayos Clínicos como Asunto , Humanos , Inmunoterapia/métodos , Inmunotoxinas/química , Leucemia/inmunología , Modelos Moleculares , Terapia Molecular Dirigida , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores
13.
J Immunother ; 37(1): 8-15, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24316551

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Compuestos de Bifenilo/farmacología , Resistencia a Antineoplásicos , Proteínas Ligadas a GPI/antagonistas & inhibidores , Inmunotoxinas/farmacología , Nitrofenoles/farmacología , Neoplasias Pancreáticas/inmunología , Sulfonamidas/farmacología , Anticuerpos Monoclonales/inmunología , Antígenos de Neoplasias/inmunología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citotoxicidad Inmunológica/efectos de los fármacos , Sinergismo Farmacológico , Proteínas Ligadas a GPI/inmunología , Humanos , Inmunotoxinas/inmunología , Mesotelina , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/terapia , Piperazinas/farmacología , Unión Proteica/inmunología , Biosíntesis de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo
14.
Mol Cancer Ther ; 13(1): 82-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24145282

RESUMEN

Recombinant immunotoxins (RIT) are agents being developed for cancer treatment. They are composed of an Fv that binds to a cancer cell, fused to a 38-kDa fragment of Pseudomonas exotoxin A. SS1P is a RIT that targets mesothelin, a protein expressed on mesothelioma as well as pancreatic, ovarian, lung, and other cancers. Because the protein tyrosine kinase family regulates a variety of cellular processes and pathways, we hypothesized that tyrosine kinases might regulate susceptibility to immunotoxin killing. To investigate their role, we used siRNAs to lower the level of expression of the 88 known tyrosine kinases. We identified five tyrosine kinases, INSR, HCK, SRC, PDGFRß, and BMX that enhance the activity of SS1P when their level of expression is lowered by siRNAs. We further investigated the Src family member HCK in this study. Knocking down of SRC slightly increased SS1P killing in A431/H9 cells, but knocking down HCK substantially enhanced killing by SS1P. We investigated the mechanism of enhancement and found that HCK knockdown enhanced SS1P cleavage by furin and lowered levels of Mcl-1 and raised Bax. We then found that Src inhibitors mimic the stimulatory effect of HCK knockdown; both SU6656 and SKI-606 (bosutinib) enhanced immunotoxin killing of mesothelin-expressing cells by SS1P and CD22-expressing cells by HA22 (moxetumomab pasudotox). SU6656 also enhanced the antitumor effects of SS1P and HA22 in mouse xenograft tumor models. Our data suggest that the combination of immunotoxin with tyrosine kinase inhibitors may be an effective way to treat some cancers.


Asunto(s)
Inmunotoxinas/administración & dosificación , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Familia-src Quinasas/biosíntesis , Compuestos de Anilina/administración & dosificación , Animales , Anticuerpos Monoclonales , Toxinas Bacterianas/administración & dosificación , Línea Celular Tumoral , Sinergismo Farmacológico , Exotoxinas/administración & dosificación , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Indoles/administración & dosificación , Mesotelina , Ratones , Neoplasias/patología , Nitrilos/administración & dosificación , Quinolinas/administración & dosificación , Sulfonamidas/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/genética
15.
Cancer Res ; 73(7): 2281-8, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23348423

RESUMEN

SS1P is a recombinant immunotoxin composed of an antimesothelin Fv fragment fused to a truncated portion of Pseudomonas exotoxin A. SS1P targets and kills mesothelin-expressing tumors, which include mesothlioma as well as ovarian, lung, and pancreatic cancers. SS1P is currently in clinical trials in mesothelioma. Because insulin acting through the insulin receptor is a survival factor for many cancer cell lines, we explored how lowering insulin receptor level would affect the cytotoxic action of SS1P. We show here that siRNA knockdown of the insulin receptor enhanced the cytotoxic action of native Pseudomonas exotoxin and enhanced SS1P toxicity on several human cell lines, but did not affect the response to other cytotoxic agents such as TRAIL, etoposide, and cycloheximide. To determine how insulin receptor knockdown enhances SS1P action, we analyzed various steps involved in cell killing. We found that insulin receptor knockdown increases the cleavage of SS1P by furin, which allows more toxin to reach the cytosol and inactivate elongation factor 2. These findings indicate that the insulin receptor negatively regulates immunotoxin action.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Toxinas Bacterianas/farmacología , Proteínas Ligadas a GPI/antagonistas & inhibidores , Inmunotoxinas/farmacología , Mesotelioma/patología , Receptor de Insulina/metabolismo , Apoptosis , Western Blotting , Proliferación Celular , Humanos , Mesotelina , Mesotelioma/tratamiento farmacológico , Mesotelioma/metabolismo , Pseudomonas aeruginosa/efectos de los fármacos , Pseudomonas aeruginosa/genética , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Insulina/antagonistas & inhibidores , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Blood ; 121(7): 1165-74, 2013 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-23243285

RESUMEN

Immune targeting of B-cell malignancies using chimeric antigen receptors (CARs) is a promising new approach, but critical factors impacting CAR efficacy remain unclear. To test the suitability of targeting CD22 on precursor B-cell acute lymphoblastic leukemia (BCP-ALL), lymphoblasts from 111 patients with BCP-ALL were assayed for CD22 expression and all were found to be CD22-positive, with median CD22 expression levels of 3500 sites/cell. Three distinct binding domains targeting CD22 were fused to various TCR signaling domains ± an IgG heavy chain constant domain (CH2CH3) to create a series of vector constructs suitable to delineate optimal CAR configuration. CARs derived from the m971 anti-CD22 mAb, which targets a proximal CD22 epitope demonstrated superior antileukemic activity compared with those incorporating other binding domains, and addition of a 4-1BB signaling domain to CD28.CD3 constructs diminished potency, whereas increasing affinity of the anti-CD22 binding motif, and extending the CD22 binding domain away from the membrane via CH2CH3 had no effect. We conclude that second-generation m971 mAb-derived anti-CD22 CARs are promising novel therapeutics that should be tested in BCP-ALL.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T/inmunología , Lectina 2 Similar a Ig de Unión al Ácido Siálico/antagonistas & inhibidores , Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología , Animales , Línea Celular Tumoral , Citocinas/metabolismo , Citotoxicidad Inmunológica , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Proteínas Recombinantes de Fusión/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Int J Cancer ; 132(4): 978-87, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22821746

RESUMEN

Synergistic killing was achieved when Small Cell Lung Cancer (SCLC) cell lines were incubated with ABT-263 and an immunotoxin directed to the transferrin receptor. SCLC lines are variably sensitive to the BH-3 only peptide mimetic, ABT-263. To determine their sensitivity to toxin-based reagents, we incubated four representative SCLC lines with a model Pseudomonas exotoxin-based immunotoxin directed to the transferrin receptor. Remarkably in 4-of-4 lines, there was little evidence of immunotoxin-mediated cytotoxicity despite near complete inhibition of protein synthesis. However, when combinations of ABT-263 and immunotoxin were added to the ABT-263-resistant cell lines (H196 and H69AR), there was synergistic killing as evidenced by increased activation of caspase 3/7, annexin V staining, and loss of cell integrity. Synergistic killing was evident at 6 hr and correlated with loss of Mcl-1. This synergy was also noted when the closely related compound ABT-737 was combined with the same immunotoxin. To establish that the synergy seen in tissue culture could be achieved in vivo, H69AR cells were grown as tumors in nude mice and shown to be susceptible to the killing action of an immunotoxin-ABT-737 combination but not to either agent alone. When immunotoxin-ABT combinations were added to ABT-263-sensitive lines (H146 and H1417), killing was additive. Our data support combination approaches for treating ABT-263-resistant SCLC with ABT-263 and a second agent that provides synergistic killing action.


Asunto(s)
Compuestos de Anilina/uso terapéutico , Compuestos de Bifenilo/farmacología , Inmunotoxinas/uso terapéutico , Nitrofenoles/farmacología , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Compuestos de Anilina/farmacología , Animales , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Inmunotoxinas/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Receptores de Transferrina/inmunología
18.
Proc Natl Acad Sci U S A ; 109(18): 6898-903, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22509046

RESUMEN

HA22 is a recombinant immunotoxin composed of an anti-CD22 Fv fused to a portion of Pseudomonas exotoxin A. HA22 produced a high rate of complete remissions in drug-resistant hairy cell leukemia and has a lower response rate in pediatric acute lymphoblastic leukemia (ALL). To understand why patients with ALL have poorer responses, we isolated an ALL cell line that is resistant to killing by HA22. The resistance is unstable; without HA22 the cells revert to HA22 sensitivity in 4 mo. We showed that in the resistant cell line, HA22 is unable to ADP ribosylate and inactivate elongation factor-2 (EF2), owing to a low level of DPH4 mRNA and protein, which prevents diphthamide biosynthesis and renders EF2 refractory to HA22. Analysis of the promoter region of the DPH4 gene shows that the CpG island was hypomethylated in the HA22-sensitive cells, heavily methylated in the resistant cells, and reverted to low methylation in the revertant cells. Our data show that immunotoxin resistance is associated with reversible CpG island methylation and silencing of DPH4 gene transcription. Incubation of sensitive cells with the methylation inhibitor 5-azacytidine prevented the emergence of resistant cells, suggesting that this agent in combination with HA22 may be useful in the treatment of some cases of ALL.


Asunto(s)
Toxinas Bacterianas/farmacología , Metilación de ADN , Exotoxinas/farmacología , Proteínas del Choque Térmico HSP40/genética , Inmunotoxinas/farmacología , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Regiones Promotoras Genéticas , Azacitidina/farmacología , Secuencia de Bases , Línea Celular Tumoral , Islas de CpG , Metilación de ADN/efectos de los fármacos , ADN de Neoplasias/genética , ADN de Neoplasias/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Humanos , Datos de Secuencia Molecular , Factor 2 de Elongación Peptídica/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología
19.
J Clin Oncol ; 30(15): 1822-8, 2012 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-22355053

RESUMEN

PURPOSE: To conduct a phase I dose-escalation trial assessing safety and response of recombinant immunotoxin moxetumomab pasudotox (CAT-8015, HA22) in chemotherapy-resistant hairy cell leukemia (HCL). PATIENTS AND METHODS: Eligible patients had relapsed/refractory HCL after ≥ two prior therapies and required treatment because of abnormal blood counts. Patients received moxetumomab pasudotox 5 to 50 µg/kg every other day for three doses (QOD ×3), with up to 16 cycles repeating at ≥ 4-week intervals if patients did not experience disease progression or develop neutralizing antibodies. RESULTS: Twenty-eight patients were enrolled, including three patients each at 5, 10, 20, and 30 µg/kg, four patients at 40 µg/kg, and 12 patients at 50 µg/kg QOD ×3 for one to 16 cycles each (median, four cycles). Dose-limiting toxicity was not observed. Two patients had transient laboratory abnormalities consistent with grade 2 hemolytic uremic syndrome with peak creatinine of 1.53 to 1.66 mg/dL and platelet nadir of 106,000 to 120,000/µL. Drug-related toxicities in 25% to 64% of the 28 patients included (in decreasing frequency) grade 1 to 2 hypoalbuminemia, aminotransferase elevations, edema, headache, hypotension, nausea, and fatigue. Of 26 patients evaluable for immunogenicity, 10 patients (38%) made antibodies neutralizing more than 75% of the cytotoxicity of 1,000 ng/mL of immunotoxin, but this immunogenicity was rare (5%) after cycle 1. The overall response rate was 86%, with responses observed at all dose levels, and 13 patients (46%) achieved complete remission (CR). Only 1 CR lasted less than 1 year, with the median disease-free survival time not yet reached at 26 months. CONCLUSION: Moxetumomab pasudotox at doses up to 50 µg/kg QOD ×3 has activity in relapsed/refractory HCL and has a safety profile that supports further clinical development for treatment of this disease.


Asunto(s)
Toxinas Bacterianas/uso terapéutico , Exotoxinas/uso terapéutico , Inmunoterapia/métodos , Leucemia de Células Pilosas/terapia , Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología , Adulto , Anciano , Toxinas Bacterianas/efectos adversos , Supervivencia sin Enfermedad , Resistencia a Antineoplásicos , Exotoxinas/efectos adversos , Femenino , Humanos , Inmunoterapia/efectos adversos , Leucemia de Células Pilosas/inmunología , Leucemia de Células Pilosas/patología , Masculino , Persona de Mediana Edad , Factores de Tiempo , Resultado del Tratamiento
20.
Cancer Res ; 71(20): 6300-9, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21998010

RESUMEN

To enable antibodies to function as cytotoxic anticancer agents, they are modified either via attachment to protein toxins or highly potent, low-molecular-weight drugs. Such molecules, termed immunotoxins and antibody-drug conjugates, respectively, represent a second revolution in antibody-mediated cancer therapy. Thus, highly toxic compounds are delivered to the interior of cancer cells based on antibody specificity for cell-surface target antigens.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias Hematológicas/tratamiento farmacológico , Inmunotoxinas/uso terapéutico , Animales , Antígenos CD/inmunología , Antígenos CD19/inmunología , Antígenos de Diferenciación Mielomonocítica/inmunología , Antineoplásicos/inmunología , Toxinas Bacterianas/uso terapéutico , Complejo CD3/inmunología , Ensayos Clínicos como Asunto , Exotoxinas/uso terapéutico , Neoplasias Hematológicas/inmunología , Humanos , Inmunotoxinas/química , Inmunotoxinas/inmunología , Subunidad alfa del Receptor de Interleucina-3/inmunología , Ratones , Receptores de Interleucina-2/inmunología , Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología , Lectina 3 Similar a Ig de Unión al Ácido Siálico , Sindecano-1/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...