Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Calcif Tissue Int ; 98(3): 294-305, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26643175

RESUMEN

The early recruitment of inflammatory cells to sites of bone fracture and trauma is a critical determinant in successful fracture healing. Released by infiltrating inflammatory cells, myeloperoxidase (MPO) and eosinophil peroxidase (EPO) are heme-containing enzymes, whose functional involvement in bone repair has mainly been studied in the context of providing a mechanism for oxidative defense against invading microorganisms. We report here novel findings that show peroxidase enzymes have the capacity to stimulate osteoblastic cells to secrete collagen I protein and generate a mineralized extracellular matrix in vitro. Mechanistic studies conducted using cultured osteoblasts show that peroxidase enzymes stimulate collagen biosynthesis at a post-translational level in a prolyl hydroxylase-dependent manner, which does not require ascorbic acid. Our studies demonstrate that osteoblasts rapidly bind and internalize both MPO and EPO, and the catalytic activity of these peroxidase enzymes is essential to support collagen I biosynthesis and subsequent release of collagen by osteoblasts. We show that EPO is capable of regulating osteogenic gene expression and matrix mineralization in culture, suggesting that peroxidase enzymes may play an important role not only in normal bone repair, but also in the progression of pathological states where infiltrating inflammatory cells are known to deposit peroxidases.


Asunto(s)
Colágeno/biosíntesis , Regulación Enzimológica de la Expresión Génica , Osteoblastos/enzimología , Peroxidasas/metabolismo , Artroplastia de Reemplazo de Cadera , Ácido Ascórbico/química , Huesos/metabolismo , Células Cultivadas , Relación Dosis-Respuesta a Droga , Ensayo de Inmunoadsorción Enzimática , Peroxidasa del Eosinófilo/metabolismo , Matriz Extracelular/metabolismo , Femenino , Regulación de la Expresión Génica , Hemo/química , Humanos , Inflamación , Procesamiento Proteico-Postraduccional , Proteínas Recombinantes/metabolismo
2.
J Exp Clin Cancer Res ; 42(1): 280, 2023 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-37880707

RESUMEN

Pancreatic cancer is one of the most common causes of cancer-related death, and the 5-year survival rate has only improved marginally over the last decade. Late detection of the disease means that in most cases the disease has advanced locally and/or metastasized, and curative surgery is not possible. Chemotherapy is still the first-line treatment however, this has only had a modest impact in improving survival, with associated toxicities. Therefore, there is an urgent need for targeted approaches to better treat pancreatic cancer, while minimizing treatment-induced side-effects. Antibody drug conjugates (ADCs) are one treatment option that could fill this gap. Here, a monoclonal antibody is used to deliver extremely potent drugs directly to the tumor site to improve on-target killing while reducing off-target toxicity. In this paper, we review the current literature for ADC targets that have been examined in vivo for treating pancreatic cancer, summarize current and on-going clinical trials using ADCs to treat pancreatic cancer and discuss potential strategies to improve their therapeutic window.


Asunto(s)
Antineoplásicos , Inmunoconjugados , Neoplasias Pancreáticas , Humanos , Inmunoconjugados/farmacología , Inmunoconjugados/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas
3.
J Immunother Cancer ; 11(3)2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36878534

RESUMEN

BACKGROUND: Emerging evidence suggests that the mechanism of chemotherapy-induced cell death may influence the antitumor immune response in patients with cancer. Unlike immunologically silent apoptosis, pyroptosis is a lytic and inflammatory form of programmed cell death characterized by pore formation in the cell membrane and release of proinflammatory factors. Gasdermin E (GSDME) has recently gained attention after cleavage of GSDME by certain chemotherapeutics has been shown to elicit pyroptosis. This study investigated the immunomodulatory effects of a mesothelin-targeting antibody drug conjugate (ADC) in mouse models of breast and colon cancer. METHODS: The antitumor effects of the ADC were studied in EMT6 breast cancer and CT26 colon cancer syngeneic mouse models. The immunomodulatory effects of the ADC were assessed by analysis of tumor-infiltrating immune cells using flow cytometry. ADC mechanism of action was evaluated by morphology, biological assays, ADC-mediated cleavage of key effector proteins, and CRISPR/Cas9-mediated knockout (KO). Finally, the antitumor effect of ADC and Fms-like tyrosine kinase-3 ligand (Flt3L) combination therapy was evaluated in tumors expressing GSDME as well as in GSDME-silenced tumors. RESULTS: The data demonstrated that the ADC controlled tumor growth and stimulated anticancer immune responses. Investigation of the mechanism of action revealed that tubulysin, the cytotoxic payload of the ADC, induced cleavage of GSDME and elicited pyroptotic cell death in GSDME-expressing cells. Using GSDME KO, we showed that GSDME expression is critical for the effectiveness of the ADC as a monotherapy. Combining the ADC with Flt3L, a cytokine that expands dendritic cells in both lymphoid and non-lymphoid tissues, restored control of GSDME KO tumors. CONCLUSIONS: Together, these results show for the first time that tubulysin and a tubulysin containing ADC can elicit pyroptosis, and that this fiery cell death is critical for antitumor immunity and therapeutic response.


Asunto(s)
Neoplasias del Colon , Inmunoconjugados , Animales , Ratones , Piroptosis , Anticuerpos , Inmunoconjugados/farmacología , Inmunoconjugados/uso terapéutico , Neoplasias del Colon/tratamiento farmacológico , Apoptosis , Modelos Animales de Enfermedad
4.
Cancer Rep (Hoboken) ; 5(8): e1543, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-34636174

RESUMEN

BACKGROUND: Platinum-based chemotherapy and radiotherapy are standard treatments for non-small cell lung cancer, which is the commonest, most lethal cancer worldwide. As a marker of treatment-induced cancer cell death, we have developed a radiodiagnostic imaging antibody, which binds to La/SSB. La/SSB is an essential, ubiquitous ribonuclear protein, which is over expressed in cancer and plays a role in resistance to cancer therapies. AIM: In this study, we examined radiation-induced DNA double strand breaks (DSB) in lung cancer cell lines and examined whether La/SSB associated with these DSB. METHOD: Three lung cancer lines (A549, H460 and LL2) were irradiated with different X-ray doses or X-radiated with a 5 Gy dose and examined at different time-points post-irradiation for DNA DSB in the form of γ-H2AX and Rad51 foci. Using fluorescence microscopy, we examined whether La/SSB and γ-H2AX co-localise and performed proximity ligation assay (PLA) and co-immunoprecipitation to confirm the interaction of these proteins. RESULTS: We found that the radio-resistant A549 cell line compared to the radio-sensitive H460 cell line showed faster resolution of radiation-induced γ-H2AX foci over time. Conversely, we found more co-localised γ-H2AX and La/SSB foci by PLA in irradiated A549 cells. CONCLUSION: The co-localisation of La/SSB with radiation-induced DNA breaks suggests a role of La/SSB in DNA repair, however further experimentation is required to validate this.


Asunto(s)
Autoantígenos , Carcinoma de Pulmón de Células no Pequeñas , Roturas del ADN de Doble Cadena , Neoplasias Pulmonares , Ribonucleoproteínas , Autoantígenos/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/radioterapia , Línea Celular Tumoral , ADN/efectos de la radiación , Roturas del ADN de Doble Cadena/efectos de la radiación , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/radioterapia , Proteínas de Unión al ARN , Ribonucleoproteínas/genética , Antígeno SS-B
5.
Biomed Pharmacother ; 151: 113090, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35567988

RESUMEN

The Fc region of a monoclonal antibody (mAb) can play a crucial role in its biodistribution and therapeutic activity. The chimeric mAb, chDAB4 (APOMAB®), which binds to dead tumor cells after DNA-damaging anticancer treatment, has been studied pre-clinically in both diagnostic and therapeutic applications in cancer. Given that macrophages contribute to the tumor accumulation of chDAB4 and its potency as an antibody drug conjugate in vivo, we next wanted to determine whether the Fc region of the chDAB4 mAb also contributed. We found that, regardless of prior labeling with chDAB4, dead EL4 lymphoma or Lewis Lung (LL2) tumor cells were phagocytosed equally by wild-type or Fcγ knock-down macrophage cell lines. A similar result was seen with bone marrow-derived macrophages from wild-type, Fcγ knock-out (KO) and NOTAM mice that express Fcγ but lack immunoreceptor tyrosine-based activation motif (ITAM) signaling. Among EL4 tumor-bearing wild-type, Fcγ KO or NOTAM mice, no differences were observed in post-chemotherapy uptake of 89Zr-labeled chDAB4. Similarly, no differences were observed between LL2 tumor-bearing wild-type and Fcγ KO mice in post-chemotherapy uptake of 89Zr-chDAB4. Also, the post-chemotherapy activity of a chDAB4-antibody drug conjugate (ADC) directed against LL2 tumors did not differ among tumor-bearing wild-type, Fcγ KO and NOTAM mice, nor did the proportions and characteristics of the LL2 tumor immune cell infiltrates differ significantly among these mice. In conclusion, Fc-FcγR interactions are not essential for the diagnostic or therapeutic applications of chDAB4 conjugates because the tumor-associated macrophages, which engulf the chDAB4-labelled dead cells, respond to endogenous 'eat me' signals rather than depend on functional FcγR expression for phagocytosis.


Asunto(s)
Inmunoconjugados , Neoplasias , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Inmunoconjugados/farmacología , Inmunoconjugados/uso terapéutico , Ratones , Receptores de IgG/genética , Receptores de IgG/metabolismo , Distribución Tisular
6.
Mol Imaging Biol ; 23(6): 914-928, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34231102

RESUMEN

PURPOSE: Early detection of tumor treatment responses represents an unmet clinical need with no approved noninvasive methods. DAB4, or its chimeric derivative, chDAB4 (APOMAB®) is an antibody that targets the Lupus associated antigen (La/SSB). La/SSB is over-expressed in malignancy and selectively targeted by chDAB4 in cancer cells dying from DNA-damaging treatment. Therefore, chDAB4 is a unique diagnostic tool that detects dead cancer cells and thus could distinguish between treatment responsive and nonresponsive patients. PROCEDURES: In clinically relevant tumor models, mice bearing subcutaneous xenografts of human ovarian or lung cancer cell lines or intraperitoneal ovarian cancer xenografts were untreated or given chemotherapy followed 24h later by chDAB4 radiolabeled with [89Zr]ZrIV. Tumor responses were monitored using bioluminescence imaging and caliper measurements. [89Zr]Zr-chDAB4 uptake in tumor and normal tissues was measured using an Albira SI Positron-Emission Tomography (PET) imager and its biodistribution was measured using a Hidex gamma-counter. RESULTS: Tumor uptake of [89Zr]Zr-chDAB4 was detected in untreated mice, and uptake significantly increased in both human lung and ovarian tumors after chemotherapy, but not in normal tissues. CONCLUSION: Given that tumors, rather than normal tissues, were targeted after chemotherapy, these results support the clinical development of chDAB4 as a radiodiagnostic imaging agent and as a potential predictive marker of treatment response.


Asunto(s)
Cisplatino , Neoplasias Ováricas , Animales , Anticuerpos Monoclonales , Muerte Celular , Línea Celular Tumoral , Electrones , Xenoinjertos , Humanos , Pulmón/patología , Ratones , Neoplasias Ováricas/diagnóstico por imagen , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/patología , Tomografía de Emisión de Positrones/métodos , Radioisótopos , Distribución Tisular , Circonio
7.
Int J Cancer ; 127(2): 345-54, 2010 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19924813

RESUMEN

Osteosarcoma (OS) is the most common primary malignant tumor of bone in children and adolescents. In spite of successful control of the primary tumor, death from lung metastasis occurs in more than a third of patients. To investigate the efficacy of zoledronic acid (ZOL) on the development, progression and metastatic spread of OS, we used a rat model of OS, with features of the disease similar to human patients, including spontaneous metastasis to lungs. Rat OS cells were inoculated into the tibial marrow cavity of syngeneic rats. OS development was associated with osteolysis mixed with new bone formation, adjacent to the periosteum and extended into the surrounding soft tissue. Metastatic foci in the lungs formed 3-4 weeks postcancer cell transplantation. Treatment with a clinically relevant dose of ZOL was initiated 1 week after tumors were established and continued once weekly or as a single dose. ZOL preserved the integrity of both trabecular and cortical bone and reduced tumor-induced bone formation. However, the overall tumor burden at the primary site was not reduced because of the persistent growth of cancer cells in the extramedullary space, which was not affected by ZOL treatment. ZOL treatment failed to prevent the metastatic spread of OS to the lungs. These findings suggest that ZOL as a single agent protects against OS-induced bone destruction but lacks efficacy against pulmonary metastases in this rat model. ZOL may have potential value as an adjuvant therapy in patients with established OS.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Neoplasias Óseas/prevención & control , Resorción Ósea/prevención & control , Difosfonatos/farmacología , Imidazoles/farmacología , Neoplasias Pulmonares/patología , Osteosarcoma/prevención & control , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Neoplasias Óseas/patología , Resorción Ósea/etiología , Resorción Ósea/patología , Caspasas/metabolismo , Proliferación Celular/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/secundario , Masculino , Osteosarcoma/patología , Ratas , Ratas Endogámicas F344 , Ácido Zoledrónico
8.
Clin Cancer Res ; 15(10): 3451-61, 2009 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-19401351

RESUMEN

PURPOSE: To evaluate the efficacy of zoledronic acid (ZOL) against osteosarcoma (OS) growth, progression, and metastatic spread using an animal model of human OS that closely resembles the human disease. EXPERIMENTAL DESIGN: Human K-HOS or KRIB OS cells, tagged or untagged with a luciferase reporter construct, were transplanted directly into the tibial cavity of nude mice. ZOL was given as weekly, or a single dose of 100 microg/kg body weight, equivalent to the 4 mg i.v. dose used clinically. Tumor growth at the primary site and as pulmonary metastases was monitored by bioluminescence imaging and histology, and OS-induced bone destruction was measured using high-resolution micro-computed tomography. RESULTS: Mice transplanted with OS cells exhibited aberrant bone remodeling in the area of cancer cell transplantation, with areas of osteolysis mixed with extensive new bone formation extending from the cortex. ZOL administration prevented osteolysis and significantly reduced the amount of OS-induced bone formation. However, ZOL had no effect on tumor burden at the primary site. Importantly, ZOL failed to reduce lung metastasis and in some cases was associated with larger and more numerous metastatic lesions. CONCLUSIONS: Our data show that clinically relevant doses of ZOL, while protecting the bone from OS-induced bone destruction, do not inhibit primary tumor growth. Moreover, lung metastases were not reduced and may even have been promoted by this treatment, indicating that caution is required when the clinical application of the bisphosphonate class of antiresorptives is considered in OS.


Asunto(s)
Difosfonatos/farmacología , Imidazoles/farmacología , Osteoblastos/efectos de los fármacos , Osteólisis/prevención & control , Osteosarcoma/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Conservadores de la Densidad Ósea/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes/métodos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Osteoblastos/patología , Osteólisis/patología , Osteosarcoma/patología , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Tibia/efectos de los fármacos , Tibia/patología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Ácido Zoledrónico
9.
Clin Cancer Res ; 15(6): 1998-2009, 2009 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-19276263

RESUMEN

PURPOSE: Multiple myeloma is an incurable disease, for which the development of new therapeutic approaches is required. Here, we report on the efficacy of recombinant soluble Apo2L/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) to inhibit tumor progression and bone destruction in a xenogeneic model of human multiple myeloma. EXPERIMENTAL DESIGN: We established a mouse model of myeloma, in which Apo2L/TRAIL-sensitive RPMI-8226 or KMS-11 cells, tagged with a triple reporter gene construct (NES-HSV-TK/GFP/Luc), were transplanted directly into the tibial marrow cavity of nude mice. Tumor burden was monitored progressively by bioluminescence imaging and the development of myeloma-induced osteolysis was measured using high resolution in vivo micro-computed tomography. RESULTS: Tumor burden increased progressively in the tibial marrow cavity of mice transplanted with Apo2L/TRAIL-sensitive RPMI-8226 or KMS-11 cells associated with extensive osteolysis directly in the area of cancer cell transplantation. Treatment of mice with recombinant soluble Apo2L/TRAIL reduced myeloma burden in the bone marrow cavity and significantly protected against myeloma-induced osteolysis. The protective effects of Apo2L/TRAIL treatment on bone were mediated by the direct apoptotic actions of Apo2L/TRAIL on myeloma cells within the bone microenvironment. CONCLUSIONS: This is the first in vivo study that investigates the efficacy of recombinant Apo2L/TRAIL on myeloma burden within the bone microenvironment and associated myeloma-induced bone destruction. Our findings that recombinant soluble Apo2L/TRAIL reduces myeloma burden within the bone microenvironment and protects the bone from myeloma-induced bone destruction argue against an inhibitory role of osteoprotegerin in Apo2L/TRAIL-induced apoptosis in vivo and highlight the need to clinically evaluate Apo2L/TRAIL in patients with multiple myeloma.


Asunto(s)
Mieloma Múltiple/tratamiento farmacológico , Osteólisis/prevención & control , Ligando Inductor de Apoptosis Relacionado con TNF/uso terapéutico , Animales , Huesos/efectos de los fármacos , Huesos/metabolismo , Huesos/patología , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Mieloma Múltiple/patología , Trasplante de Neoplasias , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Proteínas Recombinantes/uso terapéutico , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Trasplante Heterólogo
10.
J Control Release ; 327: 779-787, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-32946876

RESUMEN

APOMAB (chDAB4) is a dead tumour cell-targeting antibody which has been used preclinically as a diagnostic agent and therapeutically as a radioimmunotherapy and antibody drug conjugate (ADC). However, little is known of the intra-tumour processing of chDAB4 when bound to dead tumour cells. In this study we examine the role of macrophages in the in vitro and in vivo processing of radiolabelled chDAB4 and a chDAB4 ADC. We found that chDAB4 binds to macrophages in vitro, resulting in the killing of macrophages when using the ADC, chDAB4-SG3249. Free drug released by the macrophage processing of chDAB4-SG3249 could result in killing of 'bystander' Lewis lung (LL2) carcinoma cells. Furthermore, macrophages phagocytosed chDAB4-bound dead LL2 cells and were killed when they phagocytosed chDAB4-SG3249-bound dead LL2 cells in vitro. In vivo, we found markedly different tumour retention of chDAB4 in the LL2 tumour model depending on whether it was radiolabelled with a residualising radionuclide (89Zr), which is retained intracellularly, or a non-residualising radionuclide (124I), which can diffuse out of the cell. This prolonged retention of 89Zr vs124I indicated intra-tumoral processing of chDAB4 in vivo. The tumour uptake of 89Zr-chDAB4 was reduced after macrophage depletion, which also reduced the efficacy of the chDAB4 ADC in vivo. This study shows that macrophages can process chDAB4 and chDAB4 ADC in vitro and shows the importance of tumour-associated macrophages in the tumour retention of chDAB4 and the efficacy of chDAB4 ADC in vivo.


Asunto(s)
Preparaciones Farmacéuticas , Macrófagos Asociados a Tumores , Anticuerpos Monoclonales , Línea Celular Tumoral , Macrófagos
11.
EJNMMI Radiopharm Chem ; 5(1): 27, 2020 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-33205364

RESUMEN

PURPOSE: The chimeric monoclonal antibody (mAb) chDAB4 (APOMAB®) targets the Lupus associated (La)/Sjögren Syndrome-B (SSB) antigen, which is over-expressed in tumors but only becomes available for antibody binding in dead tumor cells. Hence, chDAB4 may be used as a novel theranostic tool to distinguish between responders and nonresponders early after chemotherapy. Here, we aimed to ascertain which positron emitter, Zirconium-89 ([89Zr]ZrIV) or Iodine-124 ([124I]I), was best suited to label chDAB4 for post-chemotherapy PET imaging of tumor-bearing mice and to determine which of two different bifunctional chelators provided optimal tumor imaging by PET using [89Zr]ZrIV-labeled chDAB4. METHODS: C57BL/6 J mice bearing subcutaneous syngeneic tumors of EL4 lymphoma were either untreated or given chemotherapy, then administered radiolabeled chDAB4 after 24 h with its biodistribution examined using PET and organ assay. We compared chDAB4 radiolabeled with [89Zr] ZrIV or [124I] I, or [89Zr]Zr-chDAB4 using either DFO-NCS or DFOSq as a chelator. RESULTS: After chemotherapy, [89Zr]Zr-chDAB4 showed higher and prolonged mean (± SD) tumor uptake of 29.5 ± 5.9 compared to 7.8 ± 1.2 for [124I] I -chDAB4. In contrast, antibody uptake in healthy tissues was not affected. Compared to DFO-NCS, DFOSq did not result in significant differences in tumor uptake of [89Zr]Zr-chDAB4 but did alter the tumor:liver ratio in treated mice 3 days after injection in favour of DFOSq (8.0 ± 1.1) compared to DFO-NCS (4.2 ± 0.7). CONCLUSION: ImmunoPET using chDAB4 radiolabeled with residualizing [89Zr] ZrIV rather than [124I] I optimized post-chemotherapy tumor uptake. Further, PET imaging characteristics were improved by DFOSq rather than DFO-NCS. Therefore, the radionuclide/chelator combination of [89Zr] ZrIV and DFOSq is preferred for the imminent clinical evaluation of chDAB4 as a selective tumor cell death radioligand.

12.
Mol Cancer Ther ; 18(2): 335-345, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30413648

RESUMEN

Antibody-drug conjugates (ADC) have revolutionized the field of cancer therapy. ADCs combine the high specificity of tumor-targeting monoclonal antibodies with potent cytotoxic drugs, which cannot be used alone because of their high toxicity. Till date, all ADCs have either targeted cell membrane proteins on tumors or the tumor vasculature and microenvironment. Here, we investigate ADCs of APOMAB (DAB4, or its chimeric derivative, chDAB4), which is a mAb targeting the La/SSB protein, which is only accessible for binding in dying or dead cancer cells. We show that DAB4-labeled dead cells are phagocytosed by macrophages, and that the apoptotic/necrotic areas within lung tumor xenografts are bound by DAB4 and are infiltrated with macrophages. We show that only DAB4-ADCs with a cleavable linker and diffusible drug are effective in two lung cancer models, particularly when given after chemotherapy. These results are consistent with other recent studies showing that direct internalization of ADCs by target cells is not essential for ADC activity because the linker can be cleaved extracellularly or through other mechanisms. Rather than targeting a tumor cell type specific antigen, DAB4-ADCs have the advantage of targeting a common trait in most solid tumors: an excess of post-apoptotic, necrotic cells either adjacent to hypoxic tumor regions or distributed more generally after cytotoxic therapy. Consequently, any antitumor effects are solely the result of bystander killing, either through internalization of the dead, ADC-bound tumor cells by macrophages, or extracellular cleavage of the ADC in the tumor microenvironment.


Asunto(s)
Anticuerpos Monoclonales/química , Inmunoconjugados/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Macrófagos/metabolismo , Células A549 , Animales , Apoptosis , Línea Celular Tumoral , Humanos , Inmunoconjugados/química , Inmunoconjugados/farmacología , Neoplasias Pulmonares/metabolismo , Ratones , Fagocitosis , Células RAW 264.7 , Microambiente Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Cancer Res ; 66(10): 5363-70, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16707463

RESUMEN

Breast cancer is the most common carcinoma that metastasizes to bone. To examine the efficacy of recombinant soluble Apo2 ligand (Apo2L)/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) against breast cancer growth in bone, we established a mouse model in which MDA-MB-231 human breast cancer cells were transplanted directly into the marrow cavity of the tibiae of athymic nude mice producing osteolytic lesions in the area of injection. All vehicle-treated control animals developed large lesions that established in the marrow cavity, eroded the cortical bone, and invaded the surrounding soft tissue, as assessed by radiography, micro-computed tomography, and histology. In contrast, animals treated with recombinant soluble Apo2L/TRAIL showed significant conservation of the tibiae, with 85% reduction in osteolysis, 90% reduction in tumor burden, and no detectable soft tissue invasion. Tumor cells explanted from Apo2L/TRAIL-treated animals were significantly more resistant to the effects of Apo2L/TRAIL when compared with the cells explanted from the vehicle-treated control animals, suggesting that prolonged treatment with Apo2/TRAIL in vivo selects for a resistant phenotype. However, such resistance was readily reversed when Apo2L/TRAIL was used in combination with clinically relevant chemotherapeutic drugs, including taxol, etoposide, doxorubicin, cisplatin, or the histone deacetylase inhibitor suberoylanilide hydroxamic acid. These studies show for the first time that Apo2L/TRAIL can prevent breast cancer-induced bone destruction and highlight the potential of this ligand for the treatment of metastatic breast cancer in bone.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Proteínas Reguladoras de la Apoptosis/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Glicoproteínas de Membrana/farmacología , Osteólisis/prevención & control , Factor de Necrosis Tumoral alfa/farmacología , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/administración & dosificación , Neoplasias de la Mama/metabolismo , Procesos de Crecimiento Celular/efectos de los fármacos , Línea Celular Tumoral , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Femenino , Proteínas Fluorescentes Verdes/biosíntesis , Humanos , Glicoproteínas de Membrana/administración & dosificación , Ratones , Ratones Desnudos , Ligando Inductor de Apoptosis Relacionado con TNF , Factor de Necrosis Tumoral alfa/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Cancer Lett ; 386: 141-150, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27865798

RESUMEN

Bone metastases occur in over 75% of patients with advanced breast cancer and are responsible for high levels of morbidity and mortality. In this study, ex vivo expanded cytotoxic Vγ9Vδ2 T cells isolated from human peripheral blood were tested for their anti-cancer efficacy in combination with zoledronic acid (ZOL), using a mouse model of osteolytic breast cancer. In vitro, expanded Vγ9Vδ2 T cells were cytotoxic against a panel of human breast cancer cell lines, and ZOL pre-treatment further sensitised breast cancer cells to killing by Vγ9Vδ2 T cells. Vγ9Vδ2 T cells adoptively transferred into NOD/SCID mice localised to osteolytic breast cancer lesions in the bone, and multiple infusions of Vγ9Vδ2 T cells reduced tumour growth in the bone. ZOL pre-treatment potentiated the anti-cancer efficacy of Vγ9Vδ2 T cells, with mice showing further reductions in tumour burden. Mice treated with the combination also had reduced tumour burden of secondary pulmonary metastases, and decreased bone degradation. Our data suggests that adoptive transfer of Vγ9Vδ2 T cell in combination with ZOL may prove an effective immunotherapeutic approach for the treatment of breast cancer bone metastases.


Asunto(s)
Antineoplásicos/farmacología , Conservadores de la Densidad Ósea/farmacología , Neoplasias Óseas/prevención & control , Neoplasias de la Mama/terapia , Difosfonatos/farmacología , Imidazoles/farmacología , Inmunoterapia Adoptiva/métodos , Neoplasias Pulmonares/prevención & control , Osteólisis/prevención & control , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Linfocitos T Citotóxicos/trasplante , Animales , Neoplasias Óseas/inmunología , Neoplasias Óseas/secundario , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , Citotoxicidad Inmunológica , Femenino , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/secundario , Activación de Linfocitos , Ratones Endogámicos NOD , Ratones SCID , Osteólisis/inmunología , Osteólisis/patología , Fenotipo , Linfocitos T Citotóxicos/inmunología , Factores de Tiempo , Ensayos Antitumor por Modelo de Xenoinjerto , Ácido Zoledrónico
15.
Mol Cell Endocrinol ; 440: 8-15, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27836774

RESUMEN

Myeloperoxidase (MPO) and eosinophil peroxidase (EPO) are heme-containing enzymes, well known for their antimicrobial activity, are released in abundance by innate immune infiltrates at sites of inflammation and injury. We have discovered new and previously unrecognised roles for heme peroxidases in extracellular matrix biosynthesis, angiogenesis, and bone mineralisation, all of which play an essential role in skeletal integrity. In this study we used in vitro models of osteoclastogenesis to investigate the effects of heme peroxidase enzymes on osteoclast differentiation and bone resorbing activity, pertinent to skeletal development and remodelling. Receptor activator of nuclear factor kappa B-ligand (RANKL) stimulates the formation of tartate-resistant acid phosphatase (TRAP) positive multinucleated cells and increases bone resorption when cultured with human peripheral blood mononuclear cells (PBMCs) or the RAW264.7 murine monocytic cell line. When RANKL was added in combination with either MPO or EPO, a dose-dependent inhibition of osteoclast differentiation and bone resorption was observed. Notably, peroxidases had no effect on the bone resorbing activity of mature osteoclasts, suggesting that the inhibitory effect of the peroxidases was limited to osteoclast precursor cells. Mechanistically, we observed that osteoclast precursor cells readily internalize peroxidases, and inhibited the phosphorylation of JNK, p38 MAPK and ERK1/2, important signalling molecules central to osteoclastogenesis. Our findings suggest that peroxidase enzymes, like MPO and EPO, may play a fundamental role in inhibiting RANKL-induced osteoclast differentiation at inflammatory sites of bone fracture and injury. Therefore, peroxidase enzymes could be considered as potential therapeutic agents to treat osteolytic bone disease and aberrant bone resorption.


Asunto(s)
Resorción Ósea/enzimología , Resorción Ósea/patología , Diferenciación Celular , Osteoclastos/enzimología , Osteoclastos/patología , Peroxidasa/metabolismo , Animales , Endocitosis/efectos de los fármacos , Humanos , Factor Estimulante de Colonias de Macrófagos/farmacología , Ratones , Ligando RANK/farmacología , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos
16.
Int J Oncol ; 50(4): 1191-1200, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28260049

RESUMEN

Myeloperoxidase (MPO) and eosinophil peroxidase (EPO) are heme-containing enzymes, well known for their antimicrobial activity, are released in high quantities by infiltrating immune cells in breast cancer. However, the functional importance of their presence within the tumour microenvironment is unclear. We have recently described a new role for peroxidases as key regulators of fibroblast and endothelial cell functionality. In the present study, we investigate for the first time, the ability of peroxidases to promote breast cancer development and progression. Using the 4T1 syngeneic murine orthotopic breast cancer model, we examined whether increased levels of peroxidases in developing mammary tumours influences primary tumour growth and metastasis. We showed that MPO and EPO stimulation increased mammary tumour growth and enhanced lung metastases, effects that were associated with reduced tumour necrosis, increased collagen deposition and neo-vascularisation within the primary tumour. In vitro, peroxidase treatment, robustly stimulated human mammary fibroblast migration and collagen type I and type VI secretion. Mechanistically, peroxidases induced the transcription of pro-tumorigenic and metastatic MMP1, MMP3 and COX-2 genes. Taken together, these findings identify peroxidases as key contributors to cancer progression by augmenting pro-tumorigenic collagen production and angiogenesis. Importantly, this identifies inflammatory peroxidases as therapeutic targets in breast cancer therapy.


Asunto(s)
Neoplasias de la Mama/metabolismo , Peroxidasa del Eosinófilo/metabolismo , Neoplasias Pulmonares/secundario , Neovascularización Patológica/metabolismo , Peroxidasa/metabolismo , Proteínas Recombinantes/metabolismo , Microambiente Tumoral , Animales , Mama/irrigación sanguínea , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Movimiento Celular , Colágeno Tipo I/metabolismo , Colágeno Tipo IV/metabolismo , Ciclooxigenasa 2/metabolismo , Progresión de la Enfermedad , Femenino , Fibroblastos , Humanos , Neoplasias Mamarias Experimentales , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/metabolismo , Ratones , Ratones Endogámicos BALB C , Terapia Molecular Dirigida , Cultivo Primario de Células
17.
Cancer Med ; 6(9): 2164-2176, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28799237

RESUMEN

Tumor hypoxia is a major cause of treatment failure for a variety of malignancies. However, hypoxia also leads to treatment opportunities as demonstrated by the development of compounds that target regions of hypoxia within tumors. Evofosfamide is a hypoxia-activated prodrug that is created by linking the hypoxia-seeking 2-nitroimidazole moiety to the cytotoxic bromo-isophosphoramide mustard (Br-IPM). When evofosfamide is delivered to hypoxic regions of tumors, the DNA cross-linking toxin, Br-IPM, is released leading to cell death. This study assessed the anticancer efficacy of evofosfamide in combination with the Proapoptotic Receptor Agonists (PARAs) dulanermin and drozitumab against human osteosarcoma in vitro and in an intratibial murine model of osteosarcoma. Under hypoxic conditions in vitro, evofosfamide cooperated with dulanermin and drozitumab, resulting in the potentiation of cytotoxicity to osteosarcoma cells. In contrast, under the same conditions, primary human osteoblasts were resistant to treatment. Animals transplanted with osteosarcoma cells directly into their tibiae developed mixed osteosclerotic/osteolytic bone lesions and consequently developed lung metastases 3 weeks post cancer cell transplantation. Tumor burden in the bone was reduced by evofosfamide treatment alone and in combination with drozitumab and prevented osteosarcoma-induced bone destruction while also reducing the growth of pulmonary metastases. These results suggest that evofosfamide may be an attractive therapeutic agent, with strong anticancer activity alone or in combination with either drozitumab or dulanermin against osteosarcoma.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Neoplasias Óseas/tratamiento farmacológico , Nitroimidazoles/administración & dosificación , Osteosarcoma/tratamiento farmacológico , Mostazas de Fosforamida/administración & dosificación , Ligando Inductor de Apoptosis Relacionado con TNF/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Nitroimidazoles/farmacología , Mostazas de Fosforamida/farmacología , Profármacos/administración & dosificación , Profármacos/farmacología , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Cancer Med ; 5(3): 534-45, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26749324

RESUMEN

Tumor hypoxia is a major cause of treatment failure for a variety of malignancies. However, hypoxia offers treatment opportunities, exemplified by the development of compounds that target hypoxic regions within tumors. Evofosfamide (TH-302) is a prodrug created by the conjugation of 2-nitroimidazole to bromo-isophosphoramide mustard (Br-IPM). When evofosfamide is delivered to hypoxic regions, the DNA cross-linking effector, Br-IPM, is released. This study assessed the cytotoxic activity of evofosfamide in vitro and its antitumor activity against osteolytic breast cancer either alone or in combination with paclitaxel in vivo. A panel of human breast cancer cell lines were treated with evofosfamide under hypoxia and assessed for cell viability. Osteolytic MDA-MB-231-TXSA cells were transplanted into the mammary fat pad, or into tibiae of mice, allowed to establish and treated with evofosfamide, paclitaxel, or both. Tumor burden was monitored using bioluminescence, and cancer-induced bone destruction was measured using micro-CT. In vitro, evofosfamide was selectively cytotoxic under hypoxic conditions. In vivo evofosfamide was tumor suppressive as a single agent and cooperated with paclitaxel to reduce mammary tumor growth. Breast cancer cells transplanted into the tibiae of mice developed osteolytic lesions. In contrast, treatment with evofosfamide or paclitaxel resulted in a significant delay in tumor growth and an overall reduction in tumor burden in bone, whereas combined treatment resulted in a significantly greater reduction in tumor burden in the tibia of mice. Evofosfamide cooperates with paclitaxel and exhibits potent tumor suppressive activity against breast cancer growth in the mammary gland and in bone.


Asunto(s)
Antineoplásicos/administración & dosificación , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/secundario , Neoplasias de la Mama/tratamiento farmacológico , Nitroimidazoles/administración & dosificación , Paclitaxel/administración & dosificación , Mostazas de Fosforamida/administración & dosificación , Animales , Antineoplásicos/farmacología , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Nitroimidazoles/farmacología , Paclitaxel/farmacología , Mostazas de Fosforamida/farmacología , Carga Tumoral/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA