Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Immunol ; 210(1): 33-39, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36445393

RESUMEN

Follicular CD8+CXCR5+ T cells are a specialized CD8+ T cell subset with unique follicular-homing capabilities that have been reported to display effector functions in viral immunity, tumor immunity, and autoimmunity. CD8+CXCR5+ T cells exhibit B cell helper functions and express CD40L, ICOS, programmed cell death protein 1 (PD-1), and BCL-6, the transcriptional regulator of CD4+CXCR5+ T follicular helper cells and of germinal center B cells. HIV is known to be sequestered in lymphoid follicles, and CD8+CXCR5+ T cell frequency is a marker for disease severity, given that HIV-infected patients with lower numbers of circulating CD8+CXCR5+ T cells display lower CD4+ T cell counts. Likewise, several groups have reported a direct correlation between the quantity of CD8+CXCR5+ T cells and suppression of HIV viral load. In this study, we observed elevated absolute numbers of CD8+CXCR5+ and CD8+CXCR5+BCL-6+PD-1+ T cells in the blood of HIV-infected participants of the Multicenter AIDS Cohort Study. We further demonstrated in vitro that activated human CD8+CXCR5+ T cells isolated from peripheral blood and tonsil from healthy donors show increased CD40L expression and induce the production of PD ligand 1 (PD-L1)+IgG+ B cells. Moreover, absolute numbers of CD8+CXCR5+ T cells significantly and positively correlated with numbers of PD-L1+ B cells found in blood of HIV-infected individuals. Altogether, these results show that activated CD8+CXCR5+ T cells have the ability to activate B cells and increase the percentage of PD-L1+ and PD-L1+IgG+ B cells, which provides insights into the early events of B cell activation and differentiation and may play a role in disease progression and lymphomagenesis in HIV-infected individuals.


Asunto(s)
Infecciones por VIH , Receptor de Muerte Celular Programada 1 , Humanos , Receptor de Muerte Celular Programada 1/metabolismo , Antígeno B7-H1/metabolismo , Linfocitos T Colaboradores-Inductores , Ligando de CD40/metabolismo , Estudios de Cohortes , Ligandos , Linfocitos T CD8-positivos , Inmunoglobulina G/metabolismo , Receptores CXCR5
2.
Invest New Drugs ; 40(5): 905-921, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-35793039

RESUMEN

Cancer remains the second most common cause of death in the US. Due to a recurrent problem with anticancer drug resistance, there is a current need for anticancer drugs with distinct modes of action for combination drug therapy We have tested two novel piperidone compounds, named 2608 (1-dichloroacetyl - 3,5-bis(3,4-difluorobenzylidene)-4-piperidone) and 2610 (1-dichloroacetyl-3,5-bis(3,4-dichlorobenzylidene)-4-piperidone), for their potential cytotoxicity on numerous human cancer cell lines. We found that both compounds were cytotoxic for breast, pancreatic, leukemia, lymphoma, colon, and fibroblast cell lines, with a cytotoxic concentration 50% (CC50) in the low micromolar to nanomolar concentration range. Further assays focused primarily on an acute lymphoblastic lymphoma and colon cancer cell lines since they were the most sensitive and resistant to the experimental piperidones. The cell death mechanism was evaluated through assays commonly used to detect the induction of apoptosis. These assays revealed that both 2608 and 2610 induced reactive oxygen species (ROS) accumulation, mitochondrial depolarization, and activated caspase-3/7. Our findings suggest that the piperidones induced cell death via the intrinsic apoptotic pathway. Additional assays revealed that both piperidones cause cell cycle alteration in lymphoma and colon cell lines. Both piperidones elicited DNA fragmentation, as evidenced by an increment in the sub-G0/G1 subpopulation in both cell lines. Similar to other related compounds, both piperidones were found to act as proteasome inhibitors by increasing the levels of poly-ubiquitinated proteins in both lymphoma and colon cell lines. Hence, the two piperidones exhibited attractive cytotoxic properties and suitable mechanisms of action, which makes them good candidates as anticancer drugs.


Asunto(s)
Antineoplásicos , Linfoma , Piperidonas , Antineoplásicos/farmacología , Apoptosis , Línea Celular Tumoral , Humanos , Masculino , Piperidonas/farmacología , Próstata
3.
J Immunol ; 200(10): 3485-3494, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29654211

RESUMEN

The transferrin receptor 1 (TfR1) is an attractive target for Ab-mediated cancer therapy. We previously developed a mouse/human chimeric IgG3 Ab (ch128.1) targeting human TfR1, which exhibits direct in vitro cytotoxicity against certain human malignant B cells through TfR1 degradation and iron deprivation. ch128.1 also demonstrates exceptional antitumor activity against the B cell malignancy multiple myeloma (MM) in xenograft models of SCID-Beige mice bearing either disseminated ARH-77 or KMS-11 cells in an early disease setting. Interestingly, this activity is observed even against KMS-11 cells, which show no sensitivity to the direct cytotoxic activity of ch128.1 in vitro. To understand the contributions of the Fc fragment, we generated a ch128.1 mutant with impaired binding to FcγRs and to the complement component C1q, which retains binding to the neonatal Fc receptor. We now report that this mutant Ab does not show antitumor activity in these two MM models, indicating a crucial role of the Fc fragment in the antitumor activity of ch128.1, which can be attributed to effector functions (Ab-dependent cell-mediated cytotoxicity, Ab-dependent cell-mediated phagocytosis, and/or complement-dependent cytotoxicity). Interestingly, in the KMS-11 model, complement depletion does not affect protection, whereas macrophage depletion does. Consistent with this observation, we found that ch128.1 induces Ab-dependent cell-mediated cytotoxicity and Ab-dependent cell-mediated phagocytosis against KMS-11 cells in the presence of murine bone marrow-derived macrophages. Finally, we found that ch128.1 therapy effectively increases survival in a late MM disease setting. Our results suggest that macrophages play a major role in ch128.1-mediated antitumor protection in our models and that ch128.1 can be effective against human B cell malignancies such as MM.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Mieloma Múltiple/tratamiento farmacológico , Receptores de Transferrina/metabolismo , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Línea Celular Tumoral , Complemento C1q/metabolismo , Citofagocitosis/efectos de los fármacos , Femenino , Humanos , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulina G/metabolismo , Ratones , Ratones SCID , Mieloma Múltiple/metabolismo
4.
Cytokine ; 120: 220-226, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31121497

RESUMEN

An antibody-cytokine fusion protein, composed of the murine single-chain cytokine interleukin-12 (IL-12) genetically fused to a human IgG3 specific for the human tumor-associated antigen HER2/neu maintains antigen binding, cytokine bioactivity, and IL-12 heparin-binding activity. This latter property is responsible for the binding of the cytokine to glycosaminoglycans (GAGs) on the cell surface and the extracellular matrix and has been implicated in modulating IL-12 bioactivity. Previous studies indicate that the p40 subunit of human and murine IL-12 is responsible for the heparin-binding activity of this heterodimeric cytokine. In the present study we used bioinformatic analysis and site-directed mutagenesis to develop a version of the antibody-(IL-12) fusion protein without heparin-binding activity. This was accomplished by replacing the basic arginine (R) and lysine (K) residues in the cluster of amino acids 254-260 (RKKEKMK) of the murine IL-12 p40 subunit by the neutral non-polar amino acid alanine (A), generating an AAAEAMA mutant fusion protein. ELISA and flow cytometry demonstrated that the antibody fusion protein lacks heparin-binding activity but retains antigen binding. A T-cell proliferation assay showed IL-12 bioactivity in this construct. However, the IL-12 bioactivity is decreased compared to its non-mutated counterpart, which is consistent with an ancillary role of the heparin-binding site of IL-12 in modulating its activity. Thus, we have defined a cluster of amino acid residues with a crucial role in the heparin-binding activity of murine IL-12 in the context of an antibody-cytokine fusion protein.


Asunto(s)
Aminoácidos/metabolismo , Anticuerpos/metabolismo , Heparina/metabolismo , Interleucina-12/química , Interleucina-12/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Animales , Antígenos de Neoplasias/metabolismo , Sitios de Unión , Línea Celular Tumoral , Proliferación Celular , Humanos , Ratones , Unión Proteica , Receptor ErbB-2/metabolismo , Linfocitos T/citología
5.
Allergy ; 74(6): 1037-1051, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30636005

RESUMEN

The microbiota can play important roles in the development of human immunity and the establishment of immune homeostasis. Lifestyle factors including diet, hygiene, and exposure to viruses or bacteria, and medical interventions with antibiotics or anti-ulcer medications, regulate phylogenetic variability and the quality of cross talk between innate and adaptive immune cells via mucosal and skin epithelia. More recently, microbiota and their composition have been linked to protective effects for health. Imbalance, however, has been linked to immune-related diseases such as allergy and cancer, characterized by impaired, or exaggerated immune tolerance, respectively. In this AllergoOncology position paper, we focus on the increasing evidence defining the microbiota composition as a key determinant of immunity and immune tolerance, linked to the risk for the development of allergic and malignant diseases. We discuss novel insights into the role of microbiota in disease and patient responses to treatments in cancer and in allergy. These may highlight opportunities to improve patient outcomes with medical interventions supported through a restored microbiome.


Asunto(s)
Asma/inmunología , Asma/microbiología , Bacterias/metabolismo , Microbioma Gastrointestinal/inmunología , Interacciones Microbiota-Huesped/inmunología , Neoplasias/inmunología , Neoplasias/microbiología , Animales , Asma/metabolismo , Bacterias/genética , Niño , Preescolar , Dieta , Epitelio/inmunología , Epitelio/microbiología , Femenino , Humanos , Hipótesis de la Higiene , Inmunidad Celular , Lactante , Masculino , Micronutrientes , Membrana Mucosa/inmunología , Membrana Mucosa/microbiología , Neoplasias/metabolismo , Filogenia
6.
Breast Cancer Res Treat ; 158(1): 113-126, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27283835

RESUMEN

Racial disparities in breast cancer incidence and outcome are a major health care challenge. Patients in the black race group more likely present with an early onset and more aggressive disease. The occurrence of high numbers of macrophages is associated with tumor progression and poor prognosis in solid malignancies. Macrophages are observed in adipose tissues surrounding dead adipocytes in "crown-like structures" (CLS). Here we investigated whether the numbers of CD163+ tumor-associated macrophages (TAMs) and/or CD163+ CLS are associated with patient survival and whether there are significant differences across blacks, non-black Latinas, and Caucasians. Our findings confirm that race is statistically significantly associated with the numbers of TAMs and CLS in breast cancer, and demonstrate that the highest numbers of CD163+ TAM/CLS are found in black breast cancer patients. Our results reveal that the density of CD206 (M2) macrophages is a significant predictor of progression-free survival univariately and is also significant after adjusting for race and for HER2, respectively. We examined whether the high numbers of TAMs detected in tumors from black women were associated with macrophage proliferation, using the Ki-67 nuclear proliferation marker. Our results reveal that TAMs actively divide when in contact with tumor cells. There is a higher ratio of proliferating macrophages in tumors from black patients. These findings suggest that interventions based on targeting TAMs may not only benefit breast cancer patients in general but also serve as an approach to remedy racial disparity resulting in better prognosis patients from minority racial groups.


Asunto(s)
Neoplasias de la Mama/etnología , Neoplasias de la Mama/patología , Macrófagos/inmunología , Negro o Afroamericano , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Neoplasias de la Mama/inmunología , Proliferación Celular , Supervivencia sin Enfermedad , Femenino , Hispánicos o Latinos , Humanos , Lectinas Tipo C/metabolismo , Macrófagos/patología , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Pronóstico , Receptores de Superficie Celular/metabolismo , Análisis de Supervivencia , Población Blanca
7.
Curr Top Microbiol Immunol ; 388: 109-49, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25553797

RESUMEN

The success of antibody therapy in cancer is consistent with the ability of these molecules to activate immune responses against tumors. Experience in clinical applications, antibody design, and advancement in technology have enabled antibodies to be engineered with enhanced efficacy against cancer cells. This allows re-evaluation of current antibody approaches dominated by antibodies of the IgG class with a new light. Antibodies of the IgE class play a central role in allergic reactions and have many properties that may be advantageous for cancer therapy. IgE-based active and passive immunotherapeutic approaches have been shown to be effective in both in vitro and in vivo models of cancer, suggesting the potential use of these approaches in humans. Further studies on the anticancer efficacy and safety profile of these IgE-based approaches are warranted in preparation for translation toward clinical application.


Asunto(s)
Inmunoglobulina E/uso terapéutico , Neoplasias/terapia , Animales , Anticuerpos/uso terapéutico , Humanos , Inmunoglobulina E/efectos adversos , Inmunoglobulina E/fisiología , Inmunoterapia , Mucina-1/inmunología , Antígeno Prostático Específico/inmunología , Receptor ErbB-2/inmunología , Vacunación
8.
Breast Cancer Res ; 16(1): R10, 2014 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-24451168

RESUMEN

INTRODUCTION: The availability of mice transgenic for the human HER2 gene (huHER2) and prone to the development of HER2-driven mammary carcinogenesis (referred to as FVB-huHER2 mice) prompted us to study active immunopreventive strategies targeting the human HER2 molecule in a tolerant host. METHODS: FVB-huHER2 mice were vaccinated with either IL-12-adjuvanted human HER2-positive cancer cells or DNA vaccine carrying chimeric human-rat HER2 sequences. Onset and number of mammary tumors were recorded to evaluate vaccine potency. Mice sera were collected and passively transferred to xenograft-bearing mice to assess their antitumor efficacy. RESULTS: Both cell and DNA vaccines significantly delayed tumor onset, leading to about 65% tumor-free mice at 70 weeks, whereas mock-vaccinated FVB-huHER2 controls developed mammary tumors at a median age of 45 weeks. In the DNA vaccinated group, 65% of mice were still tumor-free at about 90 weeks of age. The number of mammary tumors per mouse was also significantly reduced in vaccinated mice. Vaccines broke the immunological tolerance to the huHER2 transgene, inducing both humoral and cytokine responses. The DNA vaccine mainly induced a high and sustained level of anti-huHER2 antibodies, the cell vaccine also elicited interferon (IFN)-γ production. Sera of DNA-vaccinated mice transferred to xenograft-carrying mice significantly inhibited the growth of human HER2-positive cancer cells. CONCLUSIONS: Anti-huHER2 antibodies elicited in the tolerant host exert antitumor activity.


Asunto(s)
Vacunas contra el Cáncer/inmunología , Neoplasias Mamarias Animales/inmunología , Receptor ErbB-2/inmunología , Vacunas de ADN/inmunología , Traslado Adoptivo , Animales , Anticuerpos/sangre , Formación de Anticuerpos/inmunología , Línea Celular Tumoral , Femenino , Humanos , Interferón gamma/biosíntesis , Interleucina-12/inmunología , Células MCF-7 , Neoplasias Mamarias Animales/patología , Neoplasias Mamarias Animales/prevención & control , Ratones , Ratones Transgénicos , Receptor ErbB-2/genética , Bazo/citología , Bazo/trasplante
9.
J Gene Med ; 16(1-2): 11-27, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24436117

RESUMEN

BACKGROUND: We previously developed an antibody-avidin fusion protein (ch128.1Av) specific for the human transferrin receptor 1 (TfR1; CD71) to be used as a delivery vector for cancer therapy and showed that ch128.1Av delivers the biotinylated plant toxin saporin-6 into malignant B cells. However, as a result of widespread expression of TfR1, delivery of the toxin to normal cells is a concern. Therefore, we explored the potential of a dual targeted lentiviral-mediated gene therapy strategy to restrict gene expression to malignant B cells. Targeting occurs through the use of ch128.1Av or its parental antibody without avidin (ch128.1) and through transcriptional regulation using an immunoglobulin promoter. METHODS: Flow cytometry was used to detect the expression of enhanced green fluorescent protein (EGFP) in a panel of cell lines. Cell viability after specific delivery of the therapeutic gene FCU1, a chimeric enzyme consisting of cytosine deaminase genetically fused to uracil phosphoribosyltransferse that converts the 5-fluorocytosine (5-FC) prodrug into toxic metabolites, was monitored using the MTS or WST-1 viability assay. RESULTS: We found that EGFP was specifically expressed in a panel of human malignant B-cell lines, but not in human malignant T-cell lines. EGFP expression was observed in all cell lines when a ubiquitous promoter was used. Furthermore, we show the decrease of cell viability in malignant plasma cells in the presence of 5-FC and the FCU1 gene. CONCLUSIONS: The present study demonstrates that gene expression can be restricted to malignant B cells and suggests that this dual targeted gene therapy strategy may help to circumvent the potential side effects of certain TfR1-targeted protein delivery approaches.


Asunto(s)
Antígenos CD/genética , Antígenos CD/inmunología , Linfocitos B/citología , Lentivirus/genética , Receptores de Transferrina/genética , Receptores de Transferrina/inmunología , Proteínas Recombinantes de Fusión/genética , Anticuerpos/genética , Anticuerpos/inmunología , Antígenos CD/biosíntesis , Linfocitos B/metabolismo , Línea Celular Tumoral , Citosina Desaminasa/genética , Flucitosina/metabolismo , Terapia Genética/efectos adversos , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Proteínas Fluorescentes Verdes , Células HEK293 , Humanos , Células Jurkat , Neoplasias/genética , Neoplasias/terapia , Pentosiltransferasa/genética , Profármacos/efectos adversos , Profármacos/uso terapéutico , Regiones Promotoras Genéticas , Receptores de Transferrina/biosíntesis , Linfocitos T/citología , Linfocitos T/metabolismo , Transducción Genética , Virus de la Estomatitis Vesicular Indiana/genética
10.
Biochim Biophys Acta ; 1820(3): 291-317, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21851850

RESUMEN

BACKGROUND: Traditional cancer therapy can be successful in destroying tumors, but can also cause dangerous side effects. Therefore, many targeted therapies are in development. The transferrin receptor (TfR) functions in cellular iron uptake through its interaction with transferrin. This receptor is an attractive molecule for the targeted therapy of cancer since it is upregulated on the surface of many cancer types and is efficiently internalized. This receptor can be targeted in two ways: 1) for the delivery of therapeutic molecules into malignant cells or 2) to block the natural function of the receptor leading directly to cancer cell death. SCOPE OF REVIEW: In the present article we discuss the strategies used to target the TfR for the delivery of therapeutic agents into cancer cells. We provide a summary of the vast types of anti-cancer drugs that have been delivered into cancer cells employing a variety of receptor binding molecules including Tf, anti-TfR antibodies, or TfR-binding peptides alone or in combination with carrier molecules including nanoparticles and viruses. MAJOR CONCLUSIONS: Targeting the TfR has been shown to be effective in delivering many different therapeutic agents and causing cytotoxic effects in cancer cells in vitro and in vivo. GENERAL SIGNIFICANCE: The extensive use of TfR for targeted therapy attests to the versatility of targeting this receptor for therapeutic purposes against malignant cells. More advances in this area are expected to further improve the therapeutic potential of targeting the TfR for cancer therapy leading to an increase in the number of clinical trials of molecules targeting this receptor. This article is part of a Special Issue entitled Transferrins: molecular mechanisms of iron transport and disorders.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/administración & dosificación , Terapia Molecular Dirigida/métodos , Neoplasias/tratamiento farmacológico , Receptores de Transferrina/metabolismo , Antineoplásicos/metabolismo , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Proteínas Portadoras/administración & dosificación , Proteínas Portadoras/metabolismo , Sistemas de Liberación de Medicamentos , Vectores Genéticos , Humanos , Hierro/metabolismo
11.
J Virol ; 86(7): 4024-8, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22278244

RESUMEN

Five New World (NW) arenaviruses cause human hemorrhagic fevers. Four of these arenaviruses are known to enter cells by binding human transferrin receptor 1 (hTfR1). Here we show that the fifth arenavirus, Chapare virus, similarly uses hTfR1. We also identify an anti-hTfR1 antibody, ch128.1, which efficiently inhibits entry mediated by the glycoproteins of all five viruses, as well as replication of infectious Junín virus. Our data indicate that all NW hemorrhagic fever arenaviruses utilize a common hTfR1 apical-domain epitope and suggest that therapeutic agents targeting this epitope, including ch128.1 itself, can be broadly effective in treating South American hemorrhagic fevers.


Asunto(s)
Anticuerpos/inmunología , Antígenos CD/química , Antígenos CD/inmunología , Arenavirus del Nuevo Mundo/fisiología , Regulación hacia Abajo , Fiebres Hemorrágicas Virales/virología , Receptores de Transferrina/química , Receptores de Transferrina/inmunología , Internalización del Virus , Secuencia de Aminoácidos , Animales , Antígenos CD/genética , Línea Celular , Fiebres Hemorrágicas Virales/genética , Fiebres Hemorrágicas Virales/inmunología , Humanos , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , Receptores de Transferrina/genética , Receptores Virales/química , Receptores Virales/genética , Receptores Virales/inmunología , Alineación de Secuencia
12.
BMC Cancer ; 13: 195, 2013 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-23594731

RESUMEN

BACKGROUND: Prostate cancer (PCa) is the second leading cause of cancer deaths in men in the United States. The prostate-specific antigen (PSA), often found at high levels in the serum of PCa patients, has been used as a marker for PCa detection and as a target of immunotherapy. The murine IgG1 monoclonal antibody AR47.47, specific for human PSA, has been shown to enhance antigen presentation by human dendritic cells and induce both CD4 and CD8 T-cell activation when complexed with PSA. In this study, we explored the properties of a novel mouse/human chimeric anti-PSA IgE containing the variable regions of AR47.47 as a potential therapy for PCa. Our goal was to take advantage of the unique properties of IgE in order to trigger immune activation against PCa. METHODS: Binding characteristics of the antibody were determined by ELISA and flow cytometry. In vitro degranulation was determined by the release of ß-hexosaminidase from effector cells. In vivo degranulation was monitored in human FcεRIα transgenic mice using the passive cutaneous anaphylaxis assay. These mice were also used for a vaccination study to determine the in vivo anti-cancer effects of this antibody. Significant differences in survival were determined using the Log Rank test. In vitro T-cell activation was studied using human dendritic cells and autologous T cells. RESULTS: The anti-PSA IgE, expressed in murine myeloma cells, is properly assembled and secreted, and binds the antigen and FcεRI. In addition, this antibody is capable of triggering effector cell degranulation in vitro and in vivo when artificially cross-linked, but not in the presence of the natural soluble antigen, suggesting that such an interaction will not trigger systemic anaphylaxis. Importantly, the anti-PSA IgE combined with PSA also triggers immune activation in vitro and in vivo and significantly prolongs the survival of human FcεRIα transgenic mice challenged with PSA-expressing tumors in a prophylactic vaccination setting. CONCLUSIONS: The anti-PSA IgE exhibits the expected biological properties and is capable of triggering immune activation and anti-tumor protection. Further studies on this antibody as a potential PCa therapy are warranted.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Degranulación de la Célula/efectos de los fármacos , Inmunoglobulina E/uso terapéutico , Factores Inmunológicos/uso terapéutico , Antígeno Prostático Específico/inmunología , Neoplasias de la Próstata/terapia , Receptores de IgE/inmunología , Animales , Degranulación de la Célula/inmunología , Células Dendríticas , Humanos , Inmunoglobulina E/farmacología , Estimación de Kaplan-Meier , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Linfocitos T , Vacunación
13.
Cancers (Basel) ; 15(18)2023 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-37760502

RESUMEN

Multiple myeloma (MM) is an incurable malignancy of plasma cells and the second most common hematologic malignancy in the United States. Although antibodies in clinical cancer therapy are generally of the IgG class, antibodies of the IgE class have attractive properties as cancer therapeutics, such as their high affinity for Fc receptors (FcεRs), the low serum levels of endogenous IgE allowing for less competition for FcR occupancy, and the lack of inhibitory FcRs. Importantly, the FcεRs are expressed on immune cells that elicit antibody-dependent cell-mediated cytotoxicity (ADCC), antibody-dependent cell-mediated phagocytosis (ADCP), and/or antigen presentation such as mast cells, eosinophils, macrophages, and dendritic cells. We now report the development of a fully human IgE targeting human CD38 as a potential MM therapy. We targeted CD38 given its high and uniform expression on MM cells. The novel anti-CD38 IgE, expressed in mammalian cells, is properly assembled and secreted, exhibits the correct molecular weight, binds antigen and the high affinity FcεRI, and induces degranulation of FcεRI expressing cells in vitro and also in vivo in transgenic BALB/c mice expressing human FcεRIα. Moreover, the anti-CD38 IgE induces ADCC and ADCP mediated by monocytes/macrophages against human MM cells (MM.1S). Importantly, the anti-CD38 IgE also prolongs survival in a preclinical disseminated xenograft mouse model using SCID-Beige mice and human MM.1S cells when administered with human peripheral blood mononuclear cells (PBMCs) as a source of monocyte effector cells. Our results suggest that anti-CD38 IgE may be effective in humans bearing MM and other malignancies expressing CD38.

14.
Cancers (Basel) ; 15(6)2023 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-36980702

RESUMEN

Transferrin receptor 1 (TfR1), also known as CD71, is a transmembrane protein involved in the cellular uptake of iron and the regulation of cell growth. This receptor is expressed at low levels on a variety of normal cells, but is upregulated on cells with a high rate of proliferation, including malignant cells and activated immune cells. Infection with the human immunodeficiency virus (HIV) leads to the chronic activation of B cells, resulting in high expression of TfR1, B-cell dysfunction, and ultimately the development of acquired immunodeficiency syndrome-related B-cell non-Hodgkin lymphoma (AIDS-NHL). Importantly, TfR1 expression is correlated with the stage and prognosis of NHL. Thus, it is a meaningful target for antibody-based NHL therapy. We previously developed a mouse/human chimeric IgG3 specific for TfR1 (ch128.1/IgG3) and showed that this antibody exhibits antitumor activity in an in vivo model of AIDS-NHL using NOD-SCID mice challenged intraperitoneally with 2F7 human Burkitt lymphoma (BL) cells that harbor the Epstein-Barr virus (EBV). We have also developed an IgG1 version of ch128.1 that shows significant antitumor activity in SCID-Beige mouse models of disseminated multiple myeloma, another B-cell malignancy. Here, we aim to explore the utility of ch128.1/IgG1 and its humanized version (hu128.1) in mouse models of AIDS-NHL. To accomplish this goal, we used the 2F7 cell line variant 2F7-BR44, which is more aggressive than the parental cell line and forms metastases in the brain of mice after systemic (intravenous) administration. We also used the human BL cell line JB, which in contrast to 2F7, is EBV-negative, allowing us to study both EBV-infected and non-infected NHL tumors. Treatment with ch128.1/IgG1 or hu128.1 of SCID-Beige mice challenged locally (subcutaneously) with 2F7-BR44 or JB cells results in significant antitumor activity against different stages of disease. Treatment of mice challenged systemically (intravenously) with either 2F7-BR44 or JB cells also showed significant antitumor activity, including long-term survival. Taken together, our results suggest that targeting TfR1 with antibodies, such as ch128.1/IgG1 or hu128.1, has potential as an effective therapy for AIDS-NHL.

15.
Cancer Immunol Immunother ; 61(9): 1535-46, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22193986

RESUMEN

Although most monoclonal antibodies developed for cancer therapy are of the IgG class, antibodies of the IgE class have certain properties that make them attractive as cancer therapeutics. These properties include the superior affinity for the Fc epsilon receptors (FcεRs), the low serum level of IgE that minimizes competition of endogenous IgE for FcεR occupancy, and the ability to induce a broad and vigorous immune response through the interaction with multiple cells including mast cells, basophils, monocytes, macrophages, dendritic cells, and eosinophils. Tumor-targeted IgE antibodies are expected to harness the allergic response against tumors and activate a secondary, T-cell-mediated immune response. Importantly, the IgE antibody can be used for passive immunotherapy and as an adjuvant of cancer vaccines. However, there are important limitations in the use of animal models including the fact that human IgE does not interact with rodent FcεRs and that there is a different cellular distribution of FcεRs in humans and rodents. Despite these limitations, different murine models have been used with success to evaluate the in vivo anti-cancer activity of several IgE antibodies. These models include wild-type immunocompetent animals bearing syngeneic tumors, xenograft models using immunocompromised mice bearing human tumors and reconstituted with human effector cells, and human FcεRIα transgenic mice bearing syngeneic tumors. In addition, non-human primates such as cynomolgus monkeys can be potentially used for toxicological and pharmacokinetic studies. This article describes the advantages and disadvantages of these models and their use in evaluating the in vivo properties of IgE antibodies for cancer therapy.


Asunto(s)
Inmunización Pasiva/métodos , Inmunoglobulina E/inmunología , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Cancer Immunol Immunother ; 61(7): 991-1003, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22127364

RESUMEN

Breast and ovarian cancer are two of the leading causes of cancer deaths among women in the United States. Overexpression of the HER2/neu oncoprotein has been reported in patients affected with breast and ovarian cancers, and is associated with poor prognosis. To develop a novel targeted therapy for HER2/neu expressing tumors, we have constructed a fully human IgE with the variable regions of the scFv C6MH3-B1 specific for HER2/neu. This antibody was expressed in murine myeloma cells and was properly assembled and secreted. The Fc region of this antibody triggers in vitro degranulation of rat basophilic cells expressing human FcεRI (RBL SX-38) in the presence of murine mammary carcinoma cells that express human HER2/neu (D2F2/E2), but not the shed (soluble) antigen (ECD(HER2)) alone. This IgE is also capable of inducing passive cutaneous anaphylaxis in a human FcεRIα transgenic mouse model, in the presence of a cross-linking antibody, but not in the presence of soluble ECD(HER2). Additionally, IgE enhances antigen presentation in human dendritic cells and facilitates cross-priming, suggesting that the antibody is able to stimulate a secondary T-cell anti-tumor response. Furthermore, we show that this IgE significantly prolongs survival of human FcεRIα transgenic mice bearing D2F2/E2 tumors. We also report that the anti-HER2/neu IgE is well tolerated in a preliminary study conducted in Macaca fascicularis (cynomolgus) monkeys. In summary, our results suggest that this IgE should be further explored as a potential therapeutic against HER2/neu overexpressing tumors, such as breast and ovarian cancers.


Asunto(s)
Neoplasias de la Mama/inmunología , Neoplasias de la Mama/terapia , Inmunización Pasiva/métodos , Inmunoglobulina E/farmacología , Receptor ErbB-2/inmunología , Animales , Especificidad de Anticuerpos , Neoplasias de la Mama/enzimología , Células CHO , Línea Celular Tumoral , Cricetinae , Femenino , Humanos , Inmunoglobulina E/inmunología , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Ratas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Proc Natl Acad Sci U S A ; 106(46): 19322-7, 2009 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-19880745

RESUMEN

Trypanosoma brucei, a parasitic protist with a single flagellum, is the causative agent of African sleeping sickness. Propulsion of T. brucei was long believed to be by a drill-like, helical motion. Using millisecond differential interference-contrast microscopy and analyzing image sequences of cultured procyclic-form and bloodstream-form parasites, as well as bloodstream-form cells in infected mouse blood, we find that, instead, motility of T. brucei is by the propagation of kinks, separating left-handed and right-handed helical waves. Kink-driven motility, previously encountered in prokaryotes, permits T. brucei a helical propagation mechanism while avoiding the large viscous drag associated with a net rotation of the broad end of its tapering body. Our study demonstrates that millisecond differential interference-contrast microscopy can be a useful tool for uncovering important short-time features of microorganism locomotion.


Asunto(s)
Movimiento Celular , Flagelos/fisiología , Trypanosoma brucei brucei/fisiología , Tripanosomiasis Africana/microbiología , Animales , Células Cultivadas , Ratones , Ratones Endogámicos BALB C , Microscopía/métodos , Trypanosoma brucei brucei/citología
18.
J Immunother ; 45(5): 227-230, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35467582

RESUMEN

Transferrin receptor 1 (TfR1) is a universal cancer marker and a meaningful target for antibody-based immunotherapy. We previously developed a mouse/human chimeric antibody (ch128.1/IgG1) specific for the human TfR1 and reported that treatment of SCID-Beige mice bearing disseminated human multiple myeloma (MM) cells with ch128.1/IgG1 results in significant antitumor activity in early-stage and late-stage disease. Both bortezomib and lenalidomide are Food and Drug Administration (FDA) approved therapeutics used to treat MM in combination with other agents. Since combining treatments with different mechanisms of action is an effective antitumor strategy and given the relevance of bortezomib and lenalidomide in MM therapy, we decided to explore, for the first time, the combination of bortezomib or lenalidomide treatment with ch128.1/IgG1 within the context of late-stage MM disease. We found that treatment with a single dose of ch128.1/IgG1, or multiple doses of bortezomib or lenalidomide, used as single agents, results in significant antitumor activity in SCID-Beige mice bearing late-stage disseminated human MM.1S tumors. However, this antitumor activity is superior when ch128.1/IgG1 is combined with either bortezomib or lenalidomide, showing significantly longer survival compared with any therapy used alone. These novel results suggest that the combinations of ch128.1/IgG1 and bortezomib or lenalidomide are promising strategies against MM.


Asunto(s)
Mieloma Múltiple , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Bortezomib/farmacología , Bortezomib/uso terapéutico , Comunicación , Dexametasona , Humanos , Inmunoglobulina G , Lenalidomida/uso terapéutico , Ratones , Ratones SCID , Receptores de Transferrina
19.
Biology (Basel) ; 11(6)2022 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-35741451

RESUMEN

In recent years, the thienopyrazole moiety has emerged as a pharmacologically active scaffold with antitumoral and kinase inhibitory activity. In this study, high-throughput screening of 2000 small molecules obtained from the ChemBridge DIVERset library revealed a unique thieno[2,3-c]pyrazole derivative (Tpz-1) with potent and selective cytotoxic effects on cancer cells. Compound Tpz-1 consistently induced cell death at low micromolar concentrations (0.19 µM to 2.99 µM) against a panel of 17 human cancer cell lines after 24 h, 48 h, or 72 h of exposure. Furthermore, an in vitro investigation of Tpz-1's mechanism of action revealed that Tpz-1 interfered with cell cycle progression, reduced phosphorylation of p38, CREB, Akt, and STAT3 kinases, induced hyperphosphorylation of Fgr, Hck, and ERK 1/2 kinases, and disrupted microtubules and mitotic spindle formation. These findings support the continued exploration of Tpz-1 and other thieno[2,3-c]pyrazole-based compounds as potential small-molecule anticancer agents.

20.
Nat Commun ; 13(1): 558, 2022 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-35091550

RESUMEN

Five New World mammarenaviruses (NWMs) cause life-threatening hemorrhagic fever (HF). Cellular entry by these viruses is mediated by human transferrin receptor 1 (hTfR1). Here, we demonstrate that an antibody (ch128.1/IgG1) which binds the apical domain of hTfR1, potently inhibits infection of attenuated and pathogenic NWMs in vitro. Computational docking of the antibody Fab crystal structure onto the known structure of hTfR1 shows an overlapping receptor-binding region shared by the Fab and the viral envelope glycoprotein GP1 subunit that binds hTfR1, and we demonstrate competitive inhibition of NWM GP1 binding by ch128.1/IgG1 as the principal mechanism of action. Importantly, ch128.1/IgG1 protects hTfR1-expressing transgenic mice against lethal NWM challenge. Additionally, the antibody is well-tolerated and only partially reduces ferritin uptake. Our findings provide the basis for the development of a novel, host receptor-targeted antibody therapeutic broadly applicable to the treatment of HF of NWM etiology.


Asunto(s)
Antígenos CD/metabolismo , Arenaviridae/metabolismo , Fiebre Hemorrágica Americana/metabolismo , Receptores de Transferrina/metabolismo , Proteínas del Envoltorio Viral/metabolismo , Células A549 , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , Antígenos CD/inmunología , Arenaviridae/efectos de los fármacos , Arenaviridae/fisiología , Chlorocebus aethiops , Fiebre Hemorrágica Americana/prevención & control , Fiebre Hemorrágica Americana/virología , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Virus Junin/efectos de los fármacos , Virus Junin/fisiología , Ratones Endogámicos C57BL , Ratones Transgénicos , Simulación del Acoplamiento Molecular , Unión Proteica/efectos de los fármacos , Receptores de Transferrina/antagonistas & inhibidores , Receptores de Transferrina/inmunología , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA