Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Proc Natl Acad Sci U S A ; 110(14): 5404-9, 2013 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-23493549

RESUMEN

The key role played by fucose in glycoprotein and cellular function has prompted significant research toward identifying recombinant and biochemical strategies for blocking its incorporation into proteins and membrane structures. Technologies surrounding engineered cell lines have evolved for the inhibition of in vitro fucosylation, but they are not applicable for in vivo use and drug development. To address this, we screened a panel of fucose analogues and identified 2-fluorofucose and 5-alkynylfucose derivatives that depleted cells of GDP-fucose, the substrate used by fucosyltransferases to incorporate fucose into protein and cellular glycans. The inhibitors were used in vitro to generate fucose-deficient antibodies with enhanced antibody-dependent cellular cytotoxicity activities. When given orally to mice, 2-fluorofucose inhibited fucosylation of endogenously produced antibodies, tumor xenograft membranes, and neutrophil adhesion glycans. We show that oral 2-fluorofucose treatment afforded complete protection from tumor engraftment in a syngeneic tumor vaccine model, inhibited neutrophil extravasation, and delayed the outgrowth of tumor xenografts in immune-deficient mice. The results point to several potential therapeutic applications for molecules that selectively block the endogenous generation of fucosylated glycan structures.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Vacunas contra el Cáncer/farmacología , Fucosa/farmacología , Fucosiltransferasas/antagonistas & inhibidores , Guanosina Difosfato Fucosa/metabolismo , Polisacáridos/metabolismo , Animales , Células CHO , Línea Celular Tumoral , Cromatografía Liquida , Cricetinae , Cricetulus , Diseño de Fármacos , Femenino , Fucosa/química , Humanos , Espectrometría de Masas , Ratones , Ratones Endogámicos BALB C , Estructura Molecular , Neutrófilos/metabolismo
2.
Mol Cancer Ther ; 23(1): 68-83, 2024 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-37775098

RESUMEN

Brentuximab vedotin, a CD30-directed antibody-drug conjugate (ADC), is approved for clinical use in multiple CD30-expressing lymphomas. The cytotoxic payload component of brentuximab vedotin is monomethyl auristatin E (MMAE), a highly potent microtubule-disrupting agent. Preclinical results provided here demonstrate that treatment of cancer cells with brentuximab vedotin or free MMAE leads to a catastrophic disruption of the microtubule network eliciting a robust endoplasmic reticulum (ER) stress response that culminates in the induction of the classic hallmarks of immunogenic cell death (ICD). In accordance with the induction of ICD, brentuximab vedotin-killed lymphoma cells drove innate immune cell activation in vitro and in vivo. In the "gold-standard" test of ICD, vaccination of mice with brentuximab vedotin or free MMAE-killed tumor cells protected animals from tumor rechallenge; in addition, T cells transferred from previously vaccinated animals slowed tumor growth in immunodeficient mice. Immunity acquired from killed tumor cell vaccination was further amplified by the addition of PD-1 blockade. In a humanized model of CD30+ B-cell tumors, treatment with brentuximab vedotin drove the expansion and recruitment of autologous Epstein-Barr virus-reactive CD8+ T cells potentiating the activity of anti-PD-1 therapy. Together, these data support the ability of brentuximab vedotin and MMAE to drive ICD in tumor cells resulting in the activation of antigen-presenting cells and augmented T-cell immunity. These data provide a strong rationale for the clinical combination of brentuximab vedotin and other MMAE-based ADCs with checkpoint inhibitors.


Asunto(s)
Infecciones por Virus de Epstein-Barr , Inmunoconjugados , Animales , Ratones , Brentuximab Vedotina , Muerte Celular Inmunogénica , Antígeno Ki-1 , Herpesvirus Humano 4/metabolismo , Inmunoconjugados/uso terapéutico , Microtúbulos/metabolismo
3.
Mol Cancer Ther ; 2024 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-38670552

RESUMEN

Delta-like ligand 3 (DLL3) is expressed in more than 70% of small cell lung cancers (SCLCs) and other neuroendocrine-derived tumor types. SCLC is highly aggressive and limited therapeutic options lead to poor prognosis for patients. HPN328 is a tri-specific T cell activating construct (TriTAC) consisting of three binding domains: a CD3 binder for T cell engagement, an albumin binder for half-life extension, and a DLL3 binder for tumor cell engagement. In vitro assays, rodent models and non-human primates were used to assess the activity of HPN328. HPN328 induces potent dose-dependent killing of DLL3-expressing SCLC cell lines in vitro concomitant with T cell activation and cytokine release. In an NCI-H82 xenograft model with established tumors, HPN328 treatment led to T cell recruitment and anti-tumor activity. In an immunocompetent mouse model expressing a human CD3ε epitope, mice previously treated with HPN328 withstood tumor rechallenge, demonstrating long-term anti-tumor immunity. When repeat doses were administered to cynomolgus monkeys, HPN328 was well tolerated up to 10 mg/kg. Pharmacodynamic changes, such as transient cytokine elevation, were observed, consistent with the expected mechanism of action of T cell engagers. HPN328 exhibited linear pharmacokinetic in the given dose range with a serum half-life of 78 to 187 hours, supporting weekly or less frequent administration of HPN328 in humans. Preclinical and nonclinical characterization suggests that HPN328 is a highly efficacious, safe, and novel therapeutic candidate. A phase 1/2 clinical trial is currently underway testing safety and efficacy in patients with DLL3 expressing malignancies.

4.
Bioconjug Chem ; 24(7): 1256-63, 2013 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-23808985

RESUMEN

A highly cytotoxic DNA cross-linking pyrrolobenzodiazepine (PBD) dimer with a valine-alanine dipeptide linker was conjugated to the anti-CD70 h1F6 mAb either through endogenous interchain cysteines or, site-specifically, through engineered cysteines at position 239 of the heavy chains. The h1F6239C-PBD conjugation strategy proved to be superior to interchain cysteine conjugation, affording an antibody-drug conjugate (ADC) with high uniformity in drug-loading and low levels of aggregation. In vitro cytotoxicity experiments demonstrated that the h1F6239C-PBD was potent and immunologically specific on CD70-positive renal cell carcinoma (RCC) and non-Hodgkin lymphoma (NHL) cell lines. The conjugate was resistant to drug loss in plasma and in circulation, and had a pharmacokinetic profile closely matching that of the parental h1F6239C antibody capped with N-ethylmaleimide (NEM). Evaluation in CD70-positive RCC and NHL mouse xenograft models showed pronounced antitumor activities at single or weekly doses as low as 0.1 mg/kg of ADC. The ADC was tolerated at 2.5 mg/kg. These results demonstrate that PBDs can be effectively used for antibody-targeted therapy.


Asunto(s)
Benzodiazepinas/química , Ligando CD27/química , Inmunoconjugados/farmacología , Animales , Dimerización , Diseño de Fármacos , Femenino , Semivida , Inmunoconjugados/química , Ratones , Ratones Endogámicos BALB C
5.
J Immunol ; 183(6): 3770-7, 2009 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-19710474

RESUMEN

Rheumatoid arthritis (RA) is characterized by inflammation and cellular proliferation in the synovial lining of joints that result in cartilage and bone destruction. Although the etiology of RA is unclear, activated lymphocytes and proinflammatory molecules, in particular TNF superfamily members, have been implicated in the disease pathology. A TNF superfamily member, CD70, is found on activated lymphocytes and shown to be important in memory and effector responses of lymphocytes. CD70 is expressed at high levels on chronically activated T cells in patients with autoimmune disorders, including RA. The involvement of CD70 in the progression of RA, however, remains unknown. In this study, we report effects of targeting CD70 on disease pathogenesis by using an anti-mouse CD70 Ab in a murine model of collagen-induced arthritis (CIA). In addition to blocking CD70 binding to its receptor CD27, the anti-CD70 Ab used also engages Fc-dependent effector functions including Ab-dependent cellular cytotoxicity, phagocytosis, and complement fixation. Treatment of mice with anti-CD70 Ab both before the onset or after the established disease in CIA model resulted in marked improvements in disease severity and significant reduction in the production of autoantibodies. Histopathological analyses of the joints of mice revealed a substantial reduction of inflammation, and bone and cartilage destruction in response to the anti-CD70 Ab treatment. These results uncover a novel role for CD27-CD70 interactions in the regulation of in vivo inflammatory response leading to arthritis, and provide a molecular basis to support the rationale for anti-CD70 therapy for autoimmune and inflammatory diseases.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Artritis Experimental/tratamiento farmacológico , Ligando CD27/antagonistas & inhibidores , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/antagonistas & inhibidores , Animales , Artritis Experimental/patología , Autoanticuerpos/análisis , Huesos/efectos de los fármacos , Huesos/patología , Cartílago/efectos de los fármacos , Cartílago/patología , Progresión de la Enfermedad , Inflamación/prevención & control , Artropatías , Masculino , Ratones
6.
Clin Cancer Res ; 27(5): 1452-1462, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33262134

RESUMEN

PURPOSE: Mesothelin (MSLN) is a glycophosphatidylinositol-linked tumor antigen overexpressed in a variety of malignancies, including ovarian, pancreatic, lung, and triple-negative breast cancer. Early signs of clinical efficacy with MSLN-targeting agents have validated MSLN as a promising target for therapeutic intervention, but therapies with improved efficacy are still needed to address the significant unmet medical need posed by MSLN-expressing cancers. EXPERIMENTAL DESIGN: We designed HPN536, a 53-kDa, trispecific, T-cell-activating protein-based construct, which binds to MSLN-expressing tumor cells, CD3ε on T cells, and to serum albumin. Experiments were conducted to assess the potency, activity, and half-life of HPN536 in in vitro assays, rodent models, and in nonhuman primates (NHP). RESULTS: HPN536 binds to MSLN-expressing tumor cells and to CD3ε on T cells, leading to T-cell activation and potent redirected target cell lysis. A third domain of HPN536 binds to serum albumin for extension of plasma half-life. In cynomolgus monkeys, HPN536 at doses ranging from 0.1 to 10 mg/kg demonstrated MSLN-dependent pharmacologic activity, was well tolerated, and showed pharmacokinetics in support of weekly dosing in humans. CONCLUSIONS: HPN536 is potent, is well tolerated, and exhibits extended half-life in NHPs. It is currently in phase I clinical testing in patients with MSLN-expressing malignancies (NCT03872206).


Asunto(s)
Inmunoterapia/métodos , Activación de Linfocitos/inmunología , Mesotelina/antagonistas & inhibidores , Neoplasias/tratamiento farmacológico , Anticuerpos de Dominio Único/farmacología , Linfocitos T/inmunología , Animales , Antígenos de Neoplasias/inmunología , Apoptosis , Proliferación Celular , Femenino , Humanos , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Neoplasias/inmunología , Neoplasias/metabolismo , Neoplasias/patología , Fragmentos de Péptidos/inmunología , Anticuerpos de Dominio Único/inmunología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Mol Cancer Ther ; 20(1): 109-120, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33203731

RESUMEN

T cells have a unique capability to eliminate cancer cells and fight malignancies. Cancer cells have adopted multiple immune evasion mechanisms aimed at inhibiting T cells. Dramatically improved patient outcomes have been achieved with therapies genetically reprogramming T cells, blocking T-cell inhibition by cancer cells, or transiently connecting T cells with cancer cells for redirected lysis. This last modality is based on antibody constructs that bind a surface antigen on cancer cells and an invariant component of the T-cell receptor. Although high response rates were observed with T-cell engagers specific for CD19, CD20, or BCMA in patients with hematologic cancers, the treatment of solid tumors has been less successful. Here, we developed and characterized a novel T-cell engager format, called TriTAC (for Trispecific T-cell Activating Construct). TriTACs are engineered with features to improve patient safety and solid tumor activity, including high stability, small size, flexible linkers, long serum half-life, and highly specific and potent redirected lysis. The present study establishes the structure/activity relationship of TriTACs and describes the development of HPN424, a PSMA- (FOLH1-) targeting TriTAC in clinical development for patients with metastatic castration-resistant prostate cancer.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Linfocitos T/metabolismo , Albúminas/farmacología , Animales , Antineoplásicos/sangre , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Complejo CD3/metabolismo , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Semivida , Humanos , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Macaca fascicularis , Ratones Endogámicos NOD , Ratones SCID , Neoplasias/patología , Antígeno Prostático Específico/metabolismo , Linfocitos T/efectos de los fármacos
8.
Blood ; 112(12): 4683-9, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-18216294

RESUMEN

Waldenström macroglobulinemia (WM) is a B-cell malignancy characterized by an IgM monoclonal gammopathy and bone marrow (BM) infiltration with lymphoplasmacytic cells (LPCs). Excess mast cells (MCs) are commonly present in WM, and provide growth and survival signals to LPCs through several TNF family ligands (CD40L, a proliferation-inducing ligand [APRIL], and B-lymphocyte stimulator factor [BLYS]). As part of these studies, we demonstrated that WM LPCs secrete soluble CD27 (sCD27), which is elevated in patients with WM (P < .001 vs healthy donors), and serves as a faithful marker of disease. Importantly, sCD27 stimulated expression of CD40L on 10 of 10 BM MC samples and APRIL on 4 of 10 BM MC samples obtained from patients with WM as well as on LAD2 MCs. Moreover, the SGN-70 humanized monoclonal antibody, which binds to CD70 (the receptor-ligand partner of CD27), abrogated sCD27 mediated up-regulation of CD40L and APRIL on WM MCs. Last, treatment of severe combined immunodeficiency-human (SCID-hu) mice with established WM using the SGN-70 antibody blocked disease progression in 12 of 12 mice, whereas disease progressed in all 5 untreated mice. The results of these studies demonstrate a functional role for sCD27 in WM pathogenesis, along with its utility as a surrogate marker of disease and a target in the treatment of WM.


Asunto(s)
Ligando CD27/metabolismo , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Macroglobulinemia de Waldenström/etiología , Animales , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Biomarcadores de Tumor/metabolismo , Ligando CD27/inmunología , Ligando CD27/fisiología , Estudios de Casos y Controles , Células Cultivadas , Humanos , Mastocitos/metabolismo , Mastocitos/patología , Ratones , Ratones SCID , Células Plasmáticas/metabolismo , Células Plasmáticas/patología , Unión Proteica , Carga Tumoral , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/fisiología , Macroglobulinemia de Waldenström/metabolismo , Macroglobulinemia de Waldenström/patología , Macroglobulinemia de Waldenström/terapia , Ensayos Antitumor por Modelo de Xenoinjerto
9.
J Pharmacol Exp Ther ; 330(3): 932-8, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19498104

RESUMEN

Antibody-drug conjugates (ADCs) made with auristatin antimitotic agents have shown significant preclinical and clinical oncology activity. SGN-75 is composed of the anti-CD70 antibody h1F6 conjugated to monomethylauristatin F through a noncleavable maleimidocaproyl linkage. To understand the pharmacologic basis of the activity of this ADC, its pharmacokinetics and biodistribution were evaluated in a mouse xenograft model with use of a dual-radiolabeled ADC. The concentrations of antibody, total auristatin (conjugated plus unconjugated), and unconjugated auristatin were measured simultaneously in serum, tumor, and 16 normal tissues. Serum pharmacokinetic parameters for antibody and total auristatin were similar with very little unconjugated auristatin observed, demonstrating a high degree of stability. The kinetic values in normal tissues generally tracked with serum: the first time point (1 h) had the highest antibody and total auristatin concentrations with low unconjugated auristatin concentrations, with the exception of organs expected to be involved in hepatobiliary clearance of the ADC, where total and unconjugated auristatin concentrations peaked at 4 h and then rapidly decreased. In tumors, antibody concentrations were maximal at 1 day, with total auristatin increasing until 2 days. Intratumoral unconjugated auristatin was a substantial fraction of the total auristatin and reached concentrations much higher than in normal tissues. The exposure of the tumor to total and unconjugated auristatin was tens to hundreds times higher than normal tissue exposure. The data establish the pharmacologic basis of activity of the ADC through specific tumor targeting, intratumoral auristatin retention, and ADC stability in the systemic circulation.


Asunto(s)
Aminobenzoatos/farmacología , Antineoplásicos/farmacología , Inmunotoxinas/farmacología , Oligopéptidos/farmacología , Animales , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacocinética , Semivida , Inmunotoxinas/farmacocinética , Marcaje Isotópico , Ratones , Ratones Desnudos , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Clin Lymphoma Myeloma ; 9(1): 90-3, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19362984

RESUMEN

Targeting leukocyte differentiation antigens is a validated approach to develop therapeutic agents for the treatment of cancer, autoimmunity, and inflammatory diseases. A subset of activation antigens transiently induced on leukocytes is particularly interesting because many of them are absent from normal tissues, including those of most vital organs, and therapeutic agents' targeting of such antigens is expected to impart limited toxicity. One such antigen, CD70, has recently emerged as an attractive potential drug target for the treatment of cancers. Whereas CD70 is only transiently expressed on activation T and B cells and mature dendritic cells, it is found to be aberrantly expressed on a variety of tumor cells, including Waldenström's macroglobulinemia. In this report, we discuss potential antibody-based therapeutic approaches targeting CD70 for tumor elimination where various mechanisms such as antibody effector functions, immune enhancement, blockade of paracrine growth loop, and delivery of cytotoxic payloads can be exploited to achieve efficacy. Indeed, early clinical trials with therapeutic anti-CD70 antibodies are currently in progress, and those for anti-CD70 drug conjugates will soon follow.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Ligando CD27/antagonistas & inhibidores , Macroglobulinemia de Waldenström/tratamiento farmacológico , Anticuerpos Monoclonales/inmunología , Ligando CD27/inmunología , Humanos , Macroglobulinemia de Waldenström/inmunología
11.
Clin Cancer Res ; 14(19): 6171-80, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18809969

RESUMEN

PURPOSE: The antitubulin agent monomethyl auristatin F (MMAF) induces potent antitumor effects when conjugated via protease cleavable linkers to antibodies targeting internalizing, tumor-specific cell surface antigens. Humanized 1F6 (h1F6) is a humanized monoclonal antibody targeting CD70, a member of the tumor necrosis factor family that is expressed on hematologic malignancies and carcinomas. Here, we tested h1F6-maleimidocaproyl (mc) MMAF conjugates, consisting of an uncleavable mc linker, for their ability to interfere with the growth of CD70-positive carcinomas. EXPERIMENTAL DESIGN: To evaluate the optimal drug per antibody ratio, we conjugated either four or eight MMAF molecules to the cysteines that comprise the interchain disulfides of h1F6 and determined antitumor activities in vitro and in xenografted mice. The tumor types tested included glioblastoma, patient-derived renal cell carcinoma (RCC) cell isolates, and standard RCC tumor cell lines. RESULTS: All h1F6-mcMMAF conjugates potently interfered with the growth of all carcinomas in vitro and resulted in complete responses of RCC tumors implanted orthotopically or s.c. in mice. In vitro, h1F6-mcMMAF(8) was generally more potent than h1F6-mcMMAF(4). However, h1F6-mcMMAF(4) displayed equal or better efficacy than h1F6-mcMMAF(8) when administered to tumor-bearing mice. CONCLUSIONS: We showed that h1F6-mcMMAF conjugates inhibited the growth of human carcinomas and that increased drug loading, while improving potency in vitro, did not substantially affect the pharmacodynamic and pharmacokinetic properties in vivo. Based on these findings, h1F6-mcMMAF(4), designated SGN-75, has been identified as a potential antibody-drug conjugate for clinical development.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales/química , Antineoplásicos/farmacología , Ligando CD27/química , Ligando CD27/inmunología , Oligopéptidos/farmacología , Animales , Anticuerpos Monoclonales Humanizados/química , Antineoplásicos/química , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/patología , Línea Celular Tumoral , Disulfuros/química , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Humanos , Concentración 50 Inhibidora , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/patología , Ratones , Trasplante de Neoplasias , Oligopéptidos/química , Tubulina (Proteína)/química , Moduladores de Tubulina/química
12.
Clin Cancer Res ; 14(23): 7763-72, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19047103

RESUMEN

PURPOSE: CD70 (CD27L) is a member of the tumor necrosis factor family aberrantly expressed on a number of hematologic malignancies and some carcinomas. CD70 expression on malignant cells coupled with its highly restricted expression on normal cells makes CD70 an attractive target for monoclonal antibody (mAb)-based therapies. We developed a humanized anti-CD70 antibody, SGN-70, and herein describe the antitumor activities of this mAb. EXPERIMENTAL DESIGN: CD70 expression on primary tumors was evaluated by immunohistochemical staining of Hodgkin lymphoma, non-Hodgkin lymphoma, multiple myeloma, and renal cell carcinoma tissue microarrays. The CD70-binding and cytotoxic activities of SGN-70 were tested in vitro using a number of cell-based assays. The in vivo antitumor properties of SGN-70 were tested in severe combined immunodeficient mice bearing disseminated lymphoma and multiple myeloma xenografts. Mechanism-of-action studies were conducted using SGN-70v, a variant mAb with equivalent target-binding activity but impaired Fcgamma receptor binding compared with SGN-70. RESULTS: Immunohistochemical analysis identified CD70 expression on approximately 40% of multiple myeloma isolates and confirmed CD70 expression on a high percentage of Hodgkin lymphoma Reed-Sternberg cells, non-Hodgkin lymphoma, and renal cell carcinoma tumors. SGN-70 lysed CD70+ tumor cells via Fc-dependent functions, including antibody-dependent cellular cytotoxicity and phagocytosis and complement fixation. In vivo, SGN-70 treatment significantly decreased tumor burden and prolonged survival of tumor-bearing mice. CONCLUSIONS: SGN-70 is a novel humanized IgG1 mAb undergoing clinical development for the treatment of CD70+ cancers. SGN-70 possesses Fc-dependent antibody effector functions and mediates antitumor activity in vivo.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antineoplásicos/inmunología , Ligando CD27/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Afinidad de Anticuerpos , Antineoplásicos/farmacología , Ligando CD27/metabolismo , Carcinoma de Células Renales/inmunología , Carcinoma de Células Renales/metabolismo , Humanos , Inmunohistoquímica , Neoplasias Renales/inmunología , Neoplasias Renales/metabolismo , Linfoma/inmunología , Linfoma/metabolismo , Ratones , Ratones SCID , Análisis de Matrices Tisulares , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Adv Exp Med Biol ; 647: 8-36, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19760064

RESUMEN

CD40 was originally identified as a receptor on B-cells that delivers contact-dependent T helper signals to B-cells through interaction with CD40 ligand (CD40L, CD154). The pivotal role played by CD40-CD40L interaction is illustrated by the defects in B-lineage cell development and the altered structures of secondary lymphoid tissues in patients and engineered mice deficient in CD40 or CD40L. CD40 signaling also provides critical functions in stimulating antigen presentation, priming of helper and cytotoxic T-cells and a variety of inflammatory reactions. As such, dysregulations in the CD40-CD40L costimulation pathway are prominently featured in human diseases ranging from inflammatory conditions to systemic autoimmunity and tissue-specific autoimmune diseases. Moreover, studies in CD40-expressing cancers have provided convincing evidence that the CD40-CD40L pathway regulates survival of neoplastic cells as well as presentation of tumor-associated antigens to the immune system. Extensive research has been devoted to explore CD40 and CD40L as drug targets. A number of anti-CD40L and anti-CD40 antibodies with diverse biological effects are in clinical development for treatment of cancer and autoimmune diseases. This chapter reviews the role of CD40-CD40L costimulation in disease pathogenesis, the characteristics of therapeutic agents targeting this pathway and status of their clinical development.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Enfermedades Autoinmunes/terapia , Antígenos CD40/inmunología , Ligando de CD40/inmunología , Neoplasias/terapia , Animales , Enfermedades Autoinmunes/inmunología , Humanos , Ratones , Neoplasias/inmunología
14.
Adv Exp Med Biol ; 647: 108-19, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19760069

RESUMEN

Expression of CD70, a member of the tumor necrosis factor superfamily, is restricted to activated T-and B-lymphocytes and mature dendritic cells. Binding of CD70 to its receptor, CD27, is important in priming, effector functions, differentiation and memory formation of T-cells as well as plasma and memory B-cell generation. Antibody blockade of CD70-CD27 interaction inhibits the onset of experimental autoimmune encephalomyelits and cardiac allograft rejection in mice. CD70 has been also detected on hematological tumors and on carcinomas. The highly restricted expression pattern of CD70 in normal tissues and its widespread expression in various malignancies as well as its potential role in autoimmune and inflammatory conditions makes it an attractive target for antibody-based therapeutics. This chapter provides an overview of the physiological role of CD70-CD27 interactions and discusses various approaches to target this pathway for therapeutic use in cancers and autoimmunity.


Asunto(s)
Enfermedades Autoinmunes/metabolismo , Ligando CD27/antagonistas & inhibidores , Neoplasias/terapia , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/antagonistas & inhibidores , Animales , Enfermedades Autoinmunes/terapia , Ligando CD27/metabolismo , Humanos , Ratones , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
15.
Cancer Res ; 66(4): 2328-37, 2006 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-16489038

RESUMEN

Metastatic renal cell carcinoma (RCC) is an aggressive disease refractory to most existing therapeutic modalities. Identifying new markers for disease progression and drug targets for RCC will benefit this unmet medical need. We report a subset of clear cell and papillary cell RCC aberrantly expressing the lymphocyte activation marker CD70, a member of the tumor necrosis factor superfamily. Importantly, CD70 expression was found to be maintained at the metastatic sites of RCC. Anti-CD70 antibody-drug conjugates (ADC) consisting of auristatin phenylalanine phenylenediamine (AFP) or monomethyl auristatin phenylalanine (MMAF), two novel derivatives of the anti-tubulin agent auristatin, mediated potent antigen-dependent cytotoxicity in CD70-expressing RCC cells. Cytotoxic activity of these anti-CD70 ADCs was associated with their internalization and subcellular trafficking through the endosomal-lysosomal pathway, disruption of cellular microtubule network, and G2-M phase cell cycle arrest. The efficiency of drug delivery using anti-CD70 as vehicle was illustrated by the much enhanced cytotoxicity of antibody-conjugated MMAF compared with free MMAF. Hence, ADCs targeted to CD70 can selectively recognize RCC, internalize, and reach the appropriate subcellular compartment(s) for drug release and tumor cell killing. In vitro cytotoxicity of these ADCs was confirmed in xenograft models using RCC cell lines. Our findings provide evidence that CD70 is an attractive target for antibody-based therapeutics against metastatic RCC and suggest that anti-CD70 ADCs can provide a new treatment approach for advanced RCC patients who currently have no chemotherapeutic options.


Asunto(s)
Antígenos CD/biosíntesis , Antineoplásicos/administración & dosificación , Carcinoma de Células Renales/inmunología , Inmunoconjugados/inmunología , Inmunoconjugados/farmacología , Neoplasias Renales/inmunología , Proteínas de la Membrana/biosíntesis , Factores de Necrosis Tumoral/biosíntesis , Animales , Antígenos CD/genética , Antígenos CD/inmunología , Apoptosis/efectos de los fármacos , Ligando CD27 , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/patología , Ciclo Celular/efectos de los fármacos , Humanos , Inmunohistoquímica , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/patología , Activación de Linfocitos , Proteínas de la Membrana/genética , Proteínas de la Membrana/inmunología , Ratones , Ratones Desnudos , Microtúbulos/efectos de los fármacos , Oligopéptidos/administración & dosificación , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Factores de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Mol Cancer Ther ; 17(2): 554-564, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29142066

RESUMEN

Treatment choices for acute myelogenous leukemia (AML) patients resistant to conventional chemotherapies are limited and novel therapeutic agents are needed. IL3 receptor alpha (IL3Rα, or CD123) is expressed on the majority of AML blasts, and there is evidence that its expression is increased on leukemic relative to normal hematopoietic stem cells, which makes it an attractive target for antibody-based therapy. Here, we report the generation and preclinical characterization of SGN-CD123A, an antibody-drug conjugate using the pyrrolobenzodiazepine dimer (PBD) linker and a humanized CD123 antibody with engineered cysteines for site-specific conjugation. Mechanistically, SGN-CD123A induces activation of DNA damage response pathways, cell-cycle changes, and apoptosis in AML cells. In vitro, SGN-CD123A-mediated potent cytotoxicity of 11/12 CD123+ AML cell lines and 20/23 primary samples from AML patients, including those with unfavorable cytogenetic profiles or FLT3 mutations. In vivo, SGN-CD123A treatment led to AML eradication in a disseminated disease model, remission in a subcutaneous xenograft model, and significant growth delay in a multidrug resistance xenograft model. Moreover, SGN-CD123A also resulted in durable complete remission of a patient-derived xenograft AML model. When combined with a FLT3 inhibitor quizartinib, SGN-CD123A enhanced the activity of quizartinib against two FLT3-mutated xenograft models. Overall, these data demonstrate that SGN-CD123A is a potent antileukemic agent, supporting an ongoing trial to evaluate its safety and efficacy in AML patients (NCT02848248). Mol Cancer Ther; 17(2); 554-64. ©2017 AACR.


Asunto(s)
Inmunoconjugados/farmacología , Subunidad alfa del Receptor de Interleucina-3/inmunología , Leucemia Mieloide Aguda/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/inmunología , Células CHO , Línea Celular Tumoral , Cricetulus , Humanos , Inmunoconjugados/inmunología , Leucemia Mieloide Aguda/inmunología , Ratones , Ratones SCID , Células THP-1 , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Cancer Res ; 65(18): 8331-8, 2005 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-16166310

RESUMEN

SGN-40 is a humanized IgG1 antihuman CD40 that is currently in a phase I clinical trial for the treatment of multiple myeloma. As surface CD40 expression on B-lineage cells is maintained from pro-B cells to plasma cells, SGN-40 may be applicable to treatment of other B-cell neoplasias, including non-Hodgkin's lymphoma. In this study, we examined potential in vitro and in vivo anti-B-lineage lymphoma activity of SGN-40. Recombinant SGN-40 was expressed and purified from Chinese hamster ovary cells and characterized based on binding affinity, specificity, and normal B-cell stimulation. The ability of SGN-40 to target neoplastic B cells was examined in vitro by proliferation inhibition, cytotoxicity, and antibody-dependent cell cytotoxicity assays and in vivo by human lymphoma xenograft models. Recombinant SGN-40 showed high affinity, Kd of approximately 1 nmol/L, and specific binding to CD40. Whereas SGN-40 was a weak agonist in stimulating normal B-cell proliferation in the absence of IL-4 and CD40L, it delivered potent proliferation inhibitory and apoptotic signals to, and mediated antibody-dependent cytotoxicity against, a panel of high-grade B-lymphoma lines. These in vitro antilymphoma effects were extended to disseminated and s.c. xenograft CD40 tumor models. In these xenograft models, the antitumor activity of SGN-40 was comparable with that of rituximab. The preclinical in vitro and in vivo antilymphoma activity of SGN-40 observed in this study provides a rationale for the clinical testing of SGN-40 in the treatment of CD40+ B-lineage lymphomas.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antígenos CD40/inmunología , Inmunoglobulina G/farmacología , Linfoma de Células B/terapia , Animales , Anticuerpos Monoclonales/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Apoptosis/efectos de los fármacos , Apoptosis/inmunología , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Caspasa 3 , Caspasas/metabolismo , Procesos de Crecimiento Celular/efectos de los fármacos , Procesos de Crecimiento Celular/inmunología , Línea Celular Tumoral , Activación Enzimática , Humanos , Inmunoglobulina G/inmunología , Linfoma de Células B/inmunología , Linfoma de Células B/patología , Ratones , Ratones SCID , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Mol Cancer Ther ; 16(7): 1347-1354, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28341790

RESUMEN

The primary mechanism of antibody-drug conjugates (ADC) is targeted delivery of a cytotoxic payload to tumor cells via cancer-associated membrane receptors. However, the tumor microenvironment likely plays a role in ADC penetration, distribution, and processing and thus impacts the overall antitumor activity. Here, we report on the potential contribution of Fc-FcγR interactions between ADCs and tumor-associated macrophages (TAM) to the preclinical antitumor activities of ADCs. In the CD30+ L-428 Hodgkin lymphoma model, anti-CD30-vcMMAE and a non-binding control (hIgG-vcMMAE) demonstrated similar antitumor activity as well as similar payload release in the tumors. IHC analysis revealed L-428 tumors contained highly abundant TAMs, which were confirmed to bind ADCs by IHC and flow cytometry. The infiltration of TAMs was further found to correlate with the antitumor activity of the non-binding hIgG-vcMMAE in five additional xenograft models. hIgG1V1-vcMMAE, bearing a mutation in the Fc region which ablates Fc gamma receptor (FcγR) binding, lost antitumor activity in three TAM-high xenograft models, suggesting Fc-FcγR interactions modulate the TAM-ADC interaction. Our results suggest that TAMs can contribute to ADC processing through FcγR interaction in preclinical tumor models and may represent an important additional mechanism for drug release from ADCs. Correlative studies in clinical trials will further shed light on whether TAMs play a role in patients' response to ADC therapies. Mol Cancer Ther; 16(7); 1347-54. ©2017 AACR.


Asunto(s)
Enfermedad de Hodgkin/tratamiento farmacológico , Inmunoconjugados/administración & dosificación , Receptores de IgG/inmunología , Microambiente Tumoral/inmunología , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Línea Celular Tumoral , Enfermedad de Hodgkin/inmunología , Enfermedad de Hodgkin/patología , Humanos , Inmunoconjugados/inmunología , Macrófagos/inmunología , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Cancer Res ; 76(9): 2710-9, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26921341

RESUMEN

Antibody-drug conjugates (ADC) comprise targeting antibodies armed with potent small-molecule payloads. ADCs demonstrate specific cell killing in clinic, but the basis of their antitumor activity is not fully understood. In this study, we investigated the degree to which payload release predicts ADC activity in vitro and in vivo ADCs were generated to target different receptors on the anaplastic large cell lymphoma line L-82, but delivered the same cytotoxic payload (monomethyl auristatin E, MMAE), and we found that the intracellular concentration of released MMAE correlated with in vitro ADC-mediated cytotoxicity independent of target expression or drug:antibody ratios. Intratumoral MMAE concentrations consistently correlated with the extent of tumor growth inhibition in tumor xenograft models. In addition, we developed a robust admixed tumor model consisting of CD30(+) and CD30(-) cancer cells to study how heterogeneity of target antigen expression, a phenomenon often observed in cancer specimens, affects the treatment response. CD30-targeting ADC delivering membrane permeable MMAE or pyrrolobenzodiazepine dimers demonstrated potent bystander killing of neighboring CD30(-) cells. In contrast, a less membrane permeable payload, MMAF, failed to mediate bystander killing in vivo, suggesting local diffusion and distribution of released payloads represents a potential mechanism of ADC-mediated bystander killing. Collectively, our findings establish that the biophysical properties and amount of released payloads are chief factors determining the overall ADC potency and bystander killing. Cancer Res; 76(9); 2710-9. ©2016 AACR.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Inmunoconjugados/farmacología , Oligopéptidos/farmacología , Animales , Línea Celular Tumoral , Cromatografía Liquida , Sistemas de Liberación de Medicamentos/métodos , Citometría de Flujo , Humanos , Inmunohistoquímica , Linfoma/patología , Espectrometría de Masas , Ratones , Ratones SCID , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Oncogene ; 23(46): 7734-45, 2004 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-15334068

RESUMEN

Expression array data for >3000 individual clones from two suppression subtractive hybridization libraries revealed 147 genes overexpressed in non-small-cell lung cancer (NSCLC) cell lines. Of these 147 genes, 30 genes have previously unknown cancer association and 65 genes have been associated with cancers other than NSCLC. The identification of 52 genes previously associated with NSCLC by different methodologies supports the validity of the strategy used here. Of the 147 genes, 19 have no prior named Unigene cluster designation, and are designated herein as L1 to L19. Quantitative real-time PCR and cancer profiling arrays were used as independent validation tools to confirm tumor overexpression for five of the 'L' genes in tumor cell lines and patient samples from NSCLC and other cancers. Follow-up studies for candidate NSCLC-associated genes can be useful in providing valuable insight into the etiology of lung cancer as well as providing potentially interesting diagnostic or therapeutic targets for further investigation.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Perfilación de la Expresión Génica , Neoplasias Pulmonares/genética , Hibridación de Ácido Nucleico/genética , Adenocarcinoma/genética , Carcinoma de Células Escamosas/genética , Mapeo Cromosómico , Cromosomas Humanos/genética , Marcadores Genéticos , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA