Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 118(26)2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34172583

RESUMEN

Costimulation via CD137 (4-1BB) enhances antitumor immunity mediated by cytotoxic T lymphocytes. Anti-CD137 agonist antibodies elicit mild liver inflammation in mice, and the maximum tolerated dose of Urelumab, an anti-human CD137 agonist monoclonal antibody, in the clinic was defined by liver inflammation-related side effects. A protease-activated prodrug form of the anti-mouse CD137 agonist antibody 1D8 (1D8 Probody therapeutic, Pb-Tx) was constructed and found to be selectively activated in the tumor microenvironment. This construct, which encompasses a protease-cleavable linker holding in place a peptide that masks the antigen binding site, exerted antitumor effects comparable to the unmodified antibody but did not result in liver inflammation. Moreover, it efficaciously synergized with both PD-1 blockade and adoptive T-cell therapy. Surprisingly, minimal active Pb-Tx reached tumor-draining lymph nodes, and regional lymphadenectomy did not abrogate antitumor efficacy. By contrast, S1P receptor-dependent recirculation of T cells was absolutely required for efficacy. The preferential cleavage of the anti-CD137 Pb-Tx by tumor proteases offers multiple therapeutic opportunities, including neoadjuvant therapy, as shown by experiments in which the Pb-Tx is given prior to surgery to avoid spontaneous metastases.


Asunto(s)
Antineoplásicos/toxicidad , Antineoplásicos/uso terapéutico , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Animales , Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Línea Celular Tumoral , Humanos , Inmunoterapia , Inflamación/patología , Hígado/patología , Neoplasias Pulmonares/secundario , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/patología , Ratones , Terapia Neoadyuvante , Péptido Hidrolasas/metabolismo
2.
Int J Mol Sci ; 24(21)2023 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-37958596

RESUMEN

Papain-like cysteine proteases are composed of 11 human cysteine cathepsins, originally located in the lysosomes. They exhibit broad specificity and act as endopeptidases and/or exopeptidases. Among them, only cathepsins B, H, C, and X/Z exhibit exopeptidase activity. Recently, cysteine cathepsins have been found to be present outside the lysosomes and often participate in various pathological processes. Hence, they have been considered key signalling molecules. Their potentially hazardous proteolytic activities are tightly regulated. This review aims to discuss recent advances in understanding the structural aspects of these four cathepsins, mechanisms of their zymogen activation, regulation of their activities, and functional aspects of these enzymes in neurodegeneration and cancer. Neurodegenerative effects have been evaluated, particularly in Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, multiple sclerosis, and neuropsychiatric disorders. Cysteine cathepsins also participate in tumour progression and metastasis through the overexpression and secretion of proteases, which trigger extracellular matrix degradation. To our knowledge, this is the first review to provide an in-depth analysis regarding the roles of cysteine cathepsins B, H, C, and X in neurodegenerative diseases and cancer. Further advances in understanding the functions of cysteine cathepsins in these conditions will result in the development of novel, targeted therapeutic strategies.


Asunto(s)
Proteasas de Cisteína , Neoplasias , Enfermedades Neurodegenerativas , Humanos , Cisteína/metabolismo , Catepsina B , Lisosomas/metabolismo
3.
Biol Chem ; 400(8): 965-977, 2019 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-30913028

RESUMEN

The role of proteases in cancer was originally thought to be limited to the breakdown of basement membranes and extracellular matrix (ECM), thereby promoting cancer cell invasion into surrounding normal tissues. It is now well understood that proteases play a much more complicated role in all stages of cancer progression and that not only tumor cells, but also stromal cells are an important source of proteases in the tumor microenvironment. Among all the proteolytic enzymes potentially associated with cancer, some proteases have taken on heightened importance due to their significant up-regulation and ability to participate at multiple stages of cancer progression and metastasis. In this review, we discuss some of the advances in understanding of the roles of several key proteases from different classes in the development and progression of cancer and the potential to leverage their upregulated activity for the development of novel targeted treatment strategies.

4.
Nano Lett ; 18(9): 5401-5410, 2018 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-30070485

RESUMEN

Owing to their unique physicochemical properties, nanomaterials have become a focus of multidisciplinary research efforts including investigations of their interactions with tumor cells and stromal compartment of tumor microenvironment (TME) toward the development of next-generation anticancer therapies. Here, we report that agglomerates of radially assembled Al hydroxide crumpled nanosheets exhibit anticancer activity due to their selective adsorption properties and positive charge. This effect was demonstrated in vitro by decreased proliferation and viability of tumor cells, and further confirmed in two murine cancer models. Moreover, Al hydroxide nanosheets almost completely inhibited the growth of murine melanoma in vivo in combination with a minimally effective dose of doxorubicin. Our direct molecular dynamics simulation demonstrated that Al hydroxide nanosheets can cause significant ion imbalance in the living cell perimembranous space through the selective adsorption of extracellular anionic species. This approach to TME dysregulation could lay the foundation for development of novel anticancer therapy strategies.


Asunto(s)
Hidróxido de Aluminio/farmacología , Proliferación Celular/efectos de los fármacos , Nanoestructuras/química , Neoplasias/tratamiento farmacológico , Hidróxido de Aluminio/química , Animales , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Células MCF-7 , Ratones , Simulación de Dinámica Molecular , Nanocáscaras/química , Microambiente Tumoral/efectos de los fármacos
5.
J Biol Chem ; 289(46): 31736-31750, 2014 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-25288807

RESUMEN

Stefin B (cystatin B) is an endogenous cysteine cathepsin inhibitor, and the loss-of-function mutations in the stefin B gene were reported in patients with Unverricht-Lundborg disease (EPM1). In this study we demonstrated that stefin B-deficient (StB KO) mice were significantly more sensitive to the lethal LPS-induced sepsis and secreted higher amounts of pro-inflammatory cytokines IL-1ß and IL-18 in the serum. We further showed that increased caspase-11 gene expression and better pro-inflammatory caspase-1 and -11 activation determined in StB KO bone marrow-derived macrophages resulted in enhanced IL-1ß processing. Pretreatment of macrophages with the cathepsin inhibitor E-64d did not affect secretion of IL-1ß, suggesting that the increased cathepsin activity determined in StB KO bone marrow-derived macrophages is not essential for inflammasome activation. Upon LPS stimulation, stefin B was targeted into the mitochondria, and the lack of stefin B resulted in the increased destabilization of mitochondrial membrane potential and mitochondrial superoxide generation. Collectively, our study demonstrates that the LPS-induced sepsis in StB KO mice is dependent on caspase-11 and mitochondrial reactive oxygen species but is not associated with the lysosomal destabilization and increased cathepsin activity in the cytosol.


Asunto(s)
Cistatina B/fisiología , Endotoxemia/metabolismo , Regulación de la Expresión Génica , Inflamación/metabolismo , Animales , Caspasas/metabolismo , Caspasas Iniciadoras , Escherichia coli/metabolismo , Inflamasomas/metabolismo , Lipopolisacáridos , Macrófagos/citología , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Mitocondrias/metabolismo , FN-kappa B/metabolismo , Especies Reactivas de Oxígeno/metabolismo
6.
Angew Chem Int Ed Engl ; 53(38): 10077-81, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-24975267

RESUMEN

Cathepsin B (CtsB) is a lysosomal cysteine proteinase that is specifically translocated to the extracellular milieu during cancer progression. The development of a lipidated CtsB inhibitor incorporated into the envelope of a liposomal nanocarrier (LNC-NS-629) is described. Ex vivo and in vivo studies confirmed selective targeting and internalization of LNC-NS-629 by tumor and stromal cells, thus validating CtsB targeting as a highly promising approach to cancer diagnosis and treatment.


Asunto(s)
Catepsina B/antagonistas & inhibidores , Transformación Celular Neoplásica/efectos de los fármacos , Inhibidores de Cisteína Proteinasa/farmacología , Sistemas de Liberación de Medicamentos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Nanoestructuras/química , Células Madre Neoplásicas/efectos de los fármacos , Animales , Catepsina B/metabolismo , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Transformación Celular Neoplásica/patología , Inhibidores de Cisteína Proteinasa/síntesis química , Inhibidores de Cisteína Proteinasa/química , Relación Dosis-Respuesta a Droga , Doxorrubicina/química , Doxorrubicina/farmacología , Portadores de Fármacos/química , Femenino , Neoplasias Mamarias Experimentales/diagnóstico , Ratones , Estructura Molecular , Células Madre Neoplásicas/patología , Relación Estructura-Actividad
7.
Methods Mol Biol ; 2773: 59-65, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38236536

RESUMEN

To study the effect of the immunologically unimpaired microenvironment on tumor progression as well as the efficacy of therapies requiring a functioning immune system, xenograft models are not suitable due to the use of immunodeficient mice. With orthotopic congenic transplantation of tumor cells into mammary tissue, we gain more control and reproducibility regarding tumor growth, while retaining a functioning immune response. Here, we provide a protocol for isolating primary tumor cells from the MMTV-PyMT mouse model and their use in developing an orthotopic mouse model of breast cancer.


Asunto(s)
Neoplasias de la Mama , Trasplantes , Humanos , Animales , Ratones , Femenino , Reproducibilidad de los Resultados , Modelos Animales de Enfermedad , Trasplante Autólogo , Microambiente Tumoral
8.
Biochim Biophys Acta ; 1824(1): 68-88, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22024571

RESUMEN

It is more than 50 years since the lysosome was discovered. Since then its hydrolytic machinery, including proteases and other hydrolases, has been fairly well identified and characterized. Among these are the cysteine cathepsins, members of the family of papain-like cysteine proteases. They have unique reactive-site properties and an uneven tissue-specific expression pattern. In living organisms their activity is a delicate balance of expression, targeting, zymogen activation, inhibition by protein inhibitors and degradation. The specificity of their substrate binding sites, small-molecule inhibitor repertoire and crystal structures are providing new tools for research and development. Their unique reactive-site properties have made it possible to confine the targets simply by the use of appropriate reactive groups. The epoxysuccinyls still dominate the field, but now nitriles seem to be the most appropriate "warhead". The view of cysteine cathepsins as lysosomal proteases is changing as there is now clear evidence of their localization in other cellular compartments. Besides being involved in protein turnover, they build an important part of the endosomal antigen presentation. Together with the growing number of non-endosomal roles of cysteine cathepsins is growing also the knowledge of their involvement in diseases such as cancer and rheumatoid arthritis, among others. Finally, cysteine cathepsins are important regulators and signaling molecules of an unimaginable number of biological processes. The current challenge is to identify their endogenous substrates, in order to gain an insight into the mechanisms of substrate degradation and processing. In this review, some of the remarkable advances that have taken place in the past decade are presented. This article is part of a Special Issue entitled: Proteolysis 50 years after the discovery of lysosome.


Asunto(s)
Catepsinas/química , Catepsinas/fisiología , Secuencia de Aminoácidos , Animales , Catepsinas/genética , Catepsinas/metabolismo , Biología Celular/tendencias , Proteasas de Cisteína/química , Proteasas de Cisteína/genética , Proteasas de Cisteína/metabolismo , Proteasas de Cisteína/fisiología , Regulación Enzimológica de la Expresión Génica/fisiología , Humanos , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
9.
Biol Chem ; 394(2): 307-16, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23152404

RESUMEN

Osteoarthritis and rheumatoid arthritis are destructive joint diseases that involve the loss of articular cartilage. Degradation of cartilage extracellular matrix is believed to occur due to imbalance between the catabolic and anabolic processes of resident chondrocytes. Previous work has suggested that various lysosomal cysteine cathepsins participate in cartilage degeneration; however, their exact roles in disease development and progression have not been elucidated. In order to study degradation processes under conditions resembling the in vivo milieu of the cartilage, we cultivated chondrocytes on a type II collagen-containing matrix. Stimulation of the cultivated chondrocytes with interleukin-1α and/or tumor necrosis factor α resulted in a time-dependent increase in cathepsin S expression and induced its secretion into the conditioned media. Using a novel bioluminescent activity-based probe, we were able to demonstrate a significant increase in proteolytic activity of cathepsin S in the conditioned media of proinflammatory cytokine-stimulated chondrocytes. For the first time, cathepsin S was demonstrated to be secreted from chondrocytes upon stimulation with the proinflammatory cytokines, and displayed proteolytic activity in culture supernatants. Its stability at neutral pH and potent proteolytic activity on extracellular matrix components mean that cathepsin S may contribute significantly to cartilage degradation and may thus be considered a potential drug target in joint diseases.


Asunto(s)
Catepsinas/biosíntesis , Catepsinas/metabolismo , Condrocitos/metabolismo , Inflamación/metabolismo , Interleucina-1alfa/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Células Cultivadas , Humanos , Proteolisis
10.
Proc Natl Acad Sci U S A ; 107(6): 2497-502, 2010 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-20133781

RESUMEN

The lysosomal cysteine proteases cathepsin B (Ctsb) and cathepsin Z (Ctsz, also called cathepsin X/P) have been implicated in cancer pathogenesis. Compensation of Ctsb by Ctsz in Ctsb (-/-) mice has been suggested. To further define the functional interplay of these proteases in the context of cancer, we generated Ctsz null mice, crossed them with Ctsb-deficient mice harboring a transgene for the mammary duct-specific expression of polyoma middle T oncogene (PymT), and analyzed the effects of single and combined Ctsb and Ctsz deficiencies on breast cancer progression. Single Ctsb deficiency resulted in delayed detection of first tumors and reduced tumor burden, whereas Ctsz-deficient mice had only a prolonged tumor-free period. However, only a trend toward reduced metastatic burden without statistical significance was detected in both single mutants. Strikingly, combined loss of Ctsb and Ctsz led to additive effects, resulting in significant and prominent delay of early and advanced tumor development, improved histopathologic tumor grading, as well as a 70% reduction in the number of lung metastases and an 80% reduction in the size of these metastases. We conclude that the double deficiency of Ctsb and Ctsz exerts significant synergistic anticancer effects, whereas the single deficiencies demonstrate at least partial reciprocal compensation.


Asunto(s)
Catepsina B/deficiencia , Catepsina Z/deficiencia , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Animales/patología , Animales , Antígenos Transformadores de Poliomavirus/genética , Antígenos Transformadores de Poliomavirus/metabolismo , Apoptosis , Catepsina B/genética , Catepsina B/metabolismo , Catepsina Z/genética , Catepsina Z/metabolismo , Movimiento Celular , Proliferación Celular , Progresión de la Enfermedad , Femenino , Técnica del Anticuerpo Fluorescente , Genotipo , Humanos , Immunoblotting , Inmunohistoquímica , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Masculino , Glándulas Mamarias Animales/metabolismo , Neoplasias Mamarias Animales/enzimología , Neoplasias Mamarias Animales/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Carga Tumoral
11.
Pharmaceutics ; 15(10)2023 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-37896224

RESUMEN

Cathepsin D is an aspartic protease and one of the most abundant proteases. It is overexpressed in many cancers and plays an important role in tumor development, progression, and metastasis. While it is a physiologically intracellular protein, cathepsin D is secreted into the extracellular matrix under pathological conditions, making it an appealing target for drug delivery systems. Here, we present the development and evaluation of a new delivery system for tumor targeting based on immunoliposomes functionalized with pepstatin A-a natural peptide inhibitor of cathepsin D. A lipid tail was added to pepstatin A, enabling its incorporation into the liposomal lipid bilayer. The successful targeting of cathepsin D was confirmed using recombinant cathepsin D and in tumor cell lines, showing the feasibility of this targeting approach and its potential for in vivo use in theragnostic applications.

12.
Nanomaterials (Basel) ; 12(4)2022 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-35214977

RESUMEN

The increasing growth in the development of various novel nanomaterials and their biomedical applications has drawn increasing attention to their biological safety and potential health impact. The most commonly used methods for nanomaterial toxicity assessment are based on laboratory experiments. In recent years, with the aid of computer modeling and data science, several in silico methods for the cytotoxicity prediction of nanomaterials have been developed. An affordable, cost-effective numerical modeling approach thus can reduce the need for in vitro and in vivo testing and predict the properties of designed or developed nanomaterials. We propose here a new in silico method for rapid cytotoxicity assessment of two-dimensional nanomaterials of arbitrary chemical composition by using free energy analysis and molecular dynamics simulations, which can be expressed by a computational indicator of nanotoxicity (CIN2D). We applied this approach to five well-known two-dimensional nanomaterials promising for biomedical applications: graphene, graphene oxide, layered double hydroxide, aloohene, and hexagonal boron nitride nanosheets. The results corroborate the available laboratory biosafety data for these nanomaterials, supporting the applicability of the developed method for predictive nanotoxicity assessment of two-dimensional nanomaterials.

13.
Cancers (Basel) ; 14(16)2022 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-36011020

RESUMEN

Nano-dimensional materials have become a focus of multiple clinical applications due to their unique physicochemical properties. Magnetic nanoparticles represent an important class of nanomaterials that are widely studied for use as magnetic resonance (MR) contrast and drug delivery agents, especially as they can be detected and manipulated remotely. Using magnetic cobalt ferrite spinel (MCFS) nanoparticles, this study was aimed at developing a multifunctional drug delivery platform with MRI capability for use in cancer treatment. We found that MCFS nanoparticles demonstrated outstanding properties for contrast MRI (r1 = 22.1 s-1mM-1 and r2 = 499 s-1mM-1) that enabled high-resolution T1- and T2-weighted MRI-based signal detection. Furthermore, MCFS nanoparticles were used for the development of a multifunctional targeted drug delivery platform for cancer treatment that is concurrently empowered with the MR contrast properties. Their therapeutic effect in systemic chemotherapy and unique MRI double-contrast properties were confirmed in vivo using a breast cancer mouse tumor model. Our study thus provides an empirical basis for the development of a novel multimodal composite drug delivery system for anticancer therapy combined with noninvasive MRI capability.

14.
Cancer Res ; 82(22): 4288-4298, 2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36112781

RESUMEN

T cell-engaging bispecific antibodies (TCB) are highly potent therapeutics that can recruit and activate cytotoxic T cells to stimulate an antitumor immune response. However, the development of TCBs against solid tumors has been limited by significant on-target toxicity to normal tissues. Probody therapeutics have been developed as a novel class of recombinant, protease-activated antibody prodrugs that are "masked" to reduce antigen binding in healthy tissues but can become conditionally unmasked by proteases that are preferentially active in the tumor microenvironment (TME). Here, we describe the preclinical efficacy and safety of CI107, a Probody TCB targeting EGFR and CD3. In vitro, the protease-activated, unmasked CI107 effectively bound EGFR and CD3 expressed on the surface of cells and induced T-cell activation, cytokine release, and cytotoxicity toward tumor cells. In contrast, dually masked CI107 displayed a >500-fold reduction in antigen binding and >15,000-fold reduction in cytotoxic activity. In vivo, CI107 potently induced dose-dependent tumor regression of established colon cancer xenografts in mice engrafted with human peripheral blood mononuclear cells. Furthermore, the MTD of CI107 in cynomolgus monkeys was more than 60-fold higher than that of the unmasked TCB, and much lower levels of toxicity were observed in animals receiving CI107. Therefore, by localizing activity to the TME and thus limiting toxicity to normal tissues, this Probody TCB demonstrates the potential to expand clinical opportunities for TCBs as effective anticancer therapies for solid tumor indications. SIGNIFICANCE: A conditionally active EGFR-CD3 T cell-engaging Probody therapeutic expands the safety window of bispecific antibodies while maintaining efficacy in preclinical solid tumor settings.


Asunto(s)
Anticuerpos Biespecíficos , Complejo CD3 , Neoplasias del Colon , Receptores ErbB , Animales , Humanos , Ratones , Anticuerpos Biespecíficos/uso terapéutico , Complejo CD3/antagonistas & inhibidores , Neoplasias del Colon/terapia , Receptores ErbB/antagonistas & inhibidores , Leucocitos Mononucleares/metabolismo , Péptido Hidrolasas/metabolismo , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Mol Cancer Ther ; 21(8): 1326-1336, 2022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35666803

RESUMEN

Probody therapeutics (Pb-Txs) are conditionally activated antibody-drug conjugates (ADCs) designed to remain inactive until proteolytically activated in the tumor microenvironment, enabling safer targeting of antigens expressed in both tumor and normal tissue. Previous attempts to target CD71, a highly expressed tumor antigen, have failed to establish an acceptable therapeutic window due to widespread normal tissue expression. This study evaluated whether a probody-drug conjugate targeting CD71 can demonstrate a favorable efficacy and tolerability profile in preclinical studies for the treatment of cancer. CX-2029, a Pb-Tx conjugated to maleimido-caproyl-valine-citrulline-p-aminobenzyloxycarbonyl-monomethyl auristatin E, was developed as a novel cancer therapeutic targeting CD71. Preclinical studies were performed to evaluate the efficacy and safety of this anti-CD71 PDC in patient-derived xenograft (PDX) mouse models and cynomolgus monkeys, respectively. CD71 expression was detected at high levels by IHC across a broad range of tumor and normal tissues. In vitro, the masked Pb-Tx form of the anti-CD71 PDC displayed a >50-fold reduced affinity for binding to CD71 on cells compared with protease-activated, unmasked anti-CD71 PDC. Potent in vivo tumor growth inhibition (stasis or regression) was observed in >80% of PDX models (28/34) at 3 or 6 mg/kg. Anti-CD71 PDC remained mostly masked (>80%) in circulation throughout dosing in cynomolgus monkeys at 2, 6, and 12 mg/kg and displayed a 10-fold improvement in tolerability compared with an anti-CD71 ADC, which was lethal. Preclinically, anti-CD71 PDC exhibits a highly efficacious and acceptable safety profile that demonstrates the utility of the Pb-Tx platform to target CD71, an otherwise undruggable target. These data support further clinical development of the anti-CD71 PDC CX-2029 as a novel cancer therapeutic.


Asunto(s)
Antineoplásicos , Inmunoconjugados , Neoplasias , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Inmunoconjugados/farmacología , Inmunoconjugados/uso terapéutico , Plomo , Macaca fascicularis/metabolismo , Ratones , Neoplasias/tratamiento farmacológico , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Clin Cancer Res ; 28(10): 2020-2029, 2022 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-35165101

RESUMEN

PURPOSE: Praluzatamab ravtansine (CX-2009) is a conditionally activated Probody drug conjugate (PDC) comprising an anti-CD166 mAb conjugated to DM4, with a protease-cleavable linker and a peptide mask that limits target engagement in normal tissue and circulation. The tumor microenvironment is enriched for proteases capable of cleaving the linker, thereby releasing the mask, allowing for localized binding of CX-2009 to CD166. CX-2009 was evaluated in a phase I/II clinical trial for patients with advanced solid tumors. PATIENTS AND METHODS: Eligible patients had metastatic cancer receiving ≥2 prior treatments. CX-2009 was administered at escalating doses every 3 weeks (0.25-10 mg/kg) or every 2 weeks (4-6 mg/kg). Primary objective was to determine the safety profile and recommended phase II dose (RP2D). RESULTS: Of 99 patients enrolled, the most prevalent subtype was breast cancer (n = 45). Median number of prior therapies was 5 (range, 1-19). Dose-limiting toxicities were observed at 8 mg/kg every 3 weeks and 6 mg/kg every 2 weeks. On the basis of tolerability, the RP2D was 7 mg/kg every 3 weeks. Tumor regressions were observed at doses ≥4 mg/kg. In the hormone receptor-positive/HER2-nonamplified breast cancer subset (n = 22), 2 patients (9%) had confirmed partial responses, and 10 patients (45%) had stable disease. Imaging with zirconium-labeled CX-2009 confirmed uptake in tumor lesions and shielding of major organs. Activated, unmasked CX-2009 was measurable in 18 of 22 posttreatment biopsies. CONCLUSIONS: CD166 is a novel, ubiquitously expressed target. CX-2009 is the first conditionally activated antibody-drug conjugate to CD166 to demonstrate both translational and clinical activity in a variety of tumor types.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Inmunoconjugados , Maitansina , Neoplasias , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Femenino , Humanos , Inmunoconjugados/efectos adversos , Maitansina/uso terapéutico , Neoplasias/patología , Microambiente Tumoral
17.
Biol Chem ; 392(11): 955-60, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21848508

RESUMEN

The development of new and effective drug delivery systems for cancer treatment represents one of the significant challenges facing biomedical technology in the last decade. Among the different methods of drug delivery, magnetic drug targeting, by enabling specific delivery of chemotherapeutic agents through the use of magnetic nanoparticles and magnetic field gradient, could be a promising approach. Recently, magnetic nanoparticles have attracted additional attention because of their potential as contrast agents for magnetic resonance imaging and heat mediators for cancer therapy. This review summarizes these approaches in the use of magnetic nanoparticles in biomedical applications and novel methods for their optimization.


Asunto(s)
Antineoplásicos/administración & dosificación , Sistemas de Liberación de Medicamentos/métodos , Imagen por Resonancia Magnética/métodos , Nanopartículas/uso terapéutico , Neoplasias/diagnóstico , Neoplasias/terapia , Animales , Medios de Contraste/química , Medios de Contraste/uso terapéutico , Humanos , Hipertermia Inducida/métodos , Campos Magnéticos , Nanopartículas/química
18.
Methods Mol Biol ; 2294: 275-293, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33742409

RESUMEN

It is becoming increasingly evident that progression and metastasis of solid cancers is driven by the interaction of oncogene-transformed cancer cells and non-malignant host cells in the tumor stroma. In this process, the immune system contributes a complex set of highly important pro- and antitumor effects, which are not readily recapitulated by commonly used xenograft cancer models in immunodeficient mice.Therefore, we provide protocols for isolation of primary tumor cells from the MMTV-PymT mouse model for metastasizing breast cancer and their resubmission to congenic immunocompetent mice by orthotopic transplantation into the mammary gland or different routes of injection to induce organ-specific experimental metastasis, including intravenous, intracardiac, and caudal artery injection of tumor cells. Moreover, we describe protocols for sensitive detection and quantification of the metastatic burden.


Asunto(s)
Adenocarcinoma/patología , Neoplasias Encefálicas/secundario , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/patología , Péptido Hidrolasas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Adenocarcinoma/enzimología , Animales , Femenino , Neoplasias Mamarias Experimentales/enzimología , Ratones , Transgenes , Células Tumorales Cultivadas
19.
Pharmaceutics ; 13(9)2021 Sep 02.
Artículo en Inglés | MEDLINE | ID: mdl-34575469

RESUMEN

Proteases are involved in the control of numerous physiological processes, and their dysregulation has been identified in a wide range of pathologies, including cancer. Protease activity is normally tightly regulated post-translationally and therefore cannot be accurately estimated based on mRNA or protein expression alone. While several types of zymography approaches to estimate protease activity exist, there remains a need for a robust and reliable technique to measure protease activity in biological tissues. We present a novel quantitative ex vivo zymography (QZ) technology based on Probody® therapeutics (Pb-Tx), a novel class of protease-activated cancer therapeutics that contain a substrate linker cleavable by tumor-associated proteases. This approach enables the measurement and comparison of protease activity in biological tissues via the detection of Pb-Tx activation. By exploiting substrate specificity and selectivity, cataloguing and differentiating protease activities is possible, with further refinement achieved using protease-specific inhibitors. Using the QZ assay and human tumor xenografts, patient tumor tissues, and patient plasma, we characterized protease activity in preclinical and clinical samples. The QZ assay offers the potential to increase our understanding of protease activity in tissues and inform diagnostic and therapeutic development for diseases, such as cancer, that are characterized by dysregulated proteolysis.

20.
Clin Cancer Res ; 27(19): 5325-5333, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34253583

RESUMEN

PURPOSE: CX-072, a PD-L1-targeting Probody therapeutic, is engineered to be activated by tumor proteases that remove a masking peptide. To study effects on biodistribution and pharmacokinetics, we performed 89Zr-CX-072 positron emission tomography (PET) imaging. EXPERIMENTAL DESIGN: Patients received ∼1 mg, 37 MBq 89Zr-CX-072 plus 0, 4, or 9 mg unlabeled CX-072 and PET scans at days 2, 4, and 7. After that, treatment comprised 10 mg/kg CX-072 q2 weeks (n = 7) + 3 mg/kg ipilimumab q3w 4× (n = 1). Normal organ tracer uptake was expressed as standardized uptake value (SUV)mean and tumor uptake as SUVmax. PD-L1 expression was measured immunohistochemically in archival tumor tissue. RESULTS: Three of the eight patients included received 10-mg protein dose resulting in a blood pool mean SUVmean ± SD of 4.27 ± 0.45 on day 4, indicating sufficient available tracer. Tumor uptake was highest at day 7, with a geometric mean SUVmax 5.89 (n = 113) and present in all patients. The median follow-up was 12 weeks (4-76+). One patient experienced stable disease and two patients a partial response. PD-L1 tumor expression was 90% in one patient and ≤1% in the other patients. Mean SUVmean ± SD day 4 at 10 mg in the spleen was 8.56 ± 1.04, bone marrow 2.21 ± 0.46, and liver 4.97 ± 0.97. Four patients out of seven showed uptake in normal lymph nodes and Waldeyer's ring. The tracer was intact in the serum or plasma. CONCLUSIONS: 89Zr-CX-072 showed tumor uptake, even in lesions with ≤1% PD-L1 expression, and modest uptake in normal lymphoid organs, with no unexpected uptake in other healthy tissues.


Asunto(s)
Neoplasias , Radioisótopos , Antígeno B7-H1/metabolismo , Humanos , Neoplasias/diagnóstico por imagen , Neoplasias/tratamiento farmacológico , Tomografía de Emisión de Positrones/métodos , Radioisótopos/uso terapéutico , Distribución Tisular , Circonio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA