Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 18(8): 889-898, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28604720

RESUMEN

Engineered crystallizable fragment (Fc) regions of antibody domains, which assume a unique and unprecedented asymmetric structure within the homodimeric Fc polypeptide, enable completely selective binding to the complement component C1q and activation of complement via the classical pathway without any concomitant engagement of the Fcγ receptor (FcγR). We used the engineered Fc domains to demonstrate in vitro and in mouse models that for therapeutic antibodies, complement-dependent cell-mediated cytotoxicity (CDCC) and complement-dependent cell-mediated phagocytosis (CDCP) by immunological effector molecules mediated the clearance of target cells with kinetics and efficacy comparable to those of the FcγR-dependent effector functions that are much better studied, while they circumvented certain adverse reactions associated with FcγR engagement. Collectively, our data highlight the importance of CDCC and CDCP in monoclonal-antibody function and provide an experimental approach for delineating the effect of complement-dependent effector-cell engagement in various therapeutic settings.


Asunto(s)
Complemento C1q/inmunología , Citotoxicidad Inmunológica/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Inmunoglobulina G/inmunología , Inmunoterapia , Neoplasias/tratamiento farmacológico , Fagocitosis/inmunología , Receptores de IgG/inmunología , Animales , Anticuerpos Monoclonales , Linfoma de Burkitt/tratamiento farmacológico , Linfoma de Burkitt/inmunología , Línea Celular Tumoral , Cromatografía en Gel , Cromatografía Liquida , Complemento C1q/metabolismo , Cristalización , Cristalografía por Rayos X , Ensayo de Inmunoadsorción Enzimática , Humanos , Fragmentos Fc de Inmunoglobulinas/metabolismo , Inmunoglobulina G/metabolismo , Técnicas In Vitro , Linfoma de Células B/tratamiento farmacológico , Linfoma de Células B/inmunología , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Linfoma de Células B Grandes Difuso/inmunología , Espectrometría de Masas , Ratones , Neoplasias/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/inmunología , Receptores de IgG/metabolismo , Resonancia por Plasmón de Superficie , Espectrometría de Masas en Tándem
2.
Blood ; 144(2): 137-144, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38643493

RESUMEN

ABSTRACT: Numerous antibody-drug conjugates (ADCs) are being developed for cancer immunotherapy. Although several of these agents have demonstrated considerable clinical efficacy and have won Food and Drug Administration (FDA) approval, in many instances, they have been characterized by adverse side effects (ASEs), which can be quite severe in a fraction of treated patients. The key hypothesis in this perspective is that many of the most serious ASEs associated with the use of ADCs in the treatment of cancer can be most readily explained and understood due to the inappropriate processing of these ADCs via pathways normally followed for immune complex clearance, which include phagocytosis and trogocytosis. We review the key published basic science experiments and clinical observations that support this idea. We propose that it is the interaction of the ADC with Fcγ receptors expressed on off-target cells and tissues that can most readily explain ADC-mediated pathologies, which therefore provides a rationale for the design of protocols to minimize ASEs. We describe measurements that should help identify those patients most likely to experience ASE due to ADC, and we propose readily available treatments as well as therapies under development for other indications that should substantially reduce ASE associated with ADC. Our focus will be on the following FDA-approved ADC for which there are substantial literatures: gemtuzumab ozogamicin and inotuzumab ozogamicin; and trastuzumab emtansine and trastuzumab deruxtecan.


Asunto(s)
Inmunoconjugados , Humanos , Inmunoconjugados/uso terapéutico , Inmunoconjugados/efectos adversos , Complejo Antígeno-Anticuerpo/inmunología , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Animales , Receptores de IgG/metabolismo , Fagocitosis/efectos de los fármacos , Antineoplásicos Inmunológicos/efectos adversos , Antineoplásicos Inmunológicos/uso terapéutico
4.
Semin Immunol ; 28(3): 309-16, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27009480

RESUMEN

Several mAbs that have been approved for the treatment of cancer make use of complement-dependent cytotoxicity (CDC) to eliminate tumor cells. Comprehensive investigations, based on in vitro studies, mouse models and analyses of patient blood samples after mAb treatment have provided key insights into the details of individual steps in the CDC reaction. Based on the lessons learned from these studies, new and innovative approaches are now being developed to increase the clinical efficacy of next generation mAbs with respect to CDC. These improvements include engineering changes in the mAbs to enhance their ability to activate complement. In addition, mAb dosing paradigms are being developed that take into account the capacity as well as the limitations of the complement system to eliminate a substantial burden of mAb-opsonized cells. Over the next few years it is likely these approaches will lead to mAbs that are far more effective in the treatment of cancer.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Proteínas del Sistema Complemento/metabolismo , Citotoxicidad Inmunológica , Inmunoterapia/métodos , Neoplasias/terapia , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos de Neoplasias/inmunología , Activación de Complemento , Ingeniería Genética , Humanos , Neoplasias/inmunología
5.
Haematologica ; 104(9): 1841-1852, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30792198

RESUMEN

CD20 monoclonal antibody therapies have significantly improved the outlook for patients with B-cell malignancies. However, many patients acquire resistance, demonstrating the need for new and improved drugs. We previously demonstrated that the natural process of antibody hexamer formation on targeted cells allows for optimal induction of complement-dependent cytotoxicity. Complement-dependent cytotoxicity can be potentiated by introducing a single point mutation such as E430G in the IgG Fc domain that enhances intermolecular Fc-Fc interactions between cell-bound IgG molecules, thereby facilitating IgG hexamer formation. Antibodies specific for CD37, a target that is abundantly expressed on healthy and malignant B cells, are generally poor inducers of complement-dependent cytotoxicity. Here we demonstrate that introduction of the hexamerization-enhancing mutation E430G in CD37-specific antibodies facilitates highly potent complement-dependent cytotoxicity in chronic lymphocytic leukemia cells ex vivo Strikingly, we observed that combinations of hexamerization-enhanced CD20 and CD37 antibodies cooperated in C1q binding and induced superior and synergistic complement-dependent cytotoxicity in patient-derived cancer cells compared to the single agents. Furthermore, CD20 and CD37 antibodies colocalized on the cell membrane, an effect that was potentiated by the hexamerization-enhancing mutation. Moreover, upon cell surface binding, CD20 and CD37 antibodies were shown to form mixed hexameric antibody complexes consisting of both antibodies each bound to their own cognate target, so-called hetero-hexamers. These findings provide novel insights into the mechanisms of synergy in antibody-mediated complement-dependent cytotoxicity and provide a rationale to explore Fc-engineering and antibody hetero-hexamerization as a tool to enhance the cooperativity and therapeutic efficacy of antibody combinations.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Antígenos CD20/inmunología , Antígenos de Neoplasias/inmunología , Proteínas del Sistema Complemento/inmunología , Fragmentos Fc de Inmunoglobulinas/inmunología , Leucemia Linfocítica Crónica de Células B/genética , Tetraspaninas/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Línea Celular Tumoral , Complemento C1q/inmunología , Transferencia Resonante de Energía de Fluorescencia , Humanos , Inmunoglobulina G/inmunología , Leucemia Linfocítica Crónica de Células B/sangre , Mutación , Unión Proteica , Rituximab/farmacología
6.
Am J Hematol ; 94(3): 327-337, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30569594

RESUMEN

Innate immune complement activation may contribute to sickle cell disease (SCD) pathogenesis. Ischemia-reperfusion physiology is a key component of the inflammatory and vaso-occlusive milieu in SCD and is associated with complement activation. C5a is an anaphylatoxin, a potent pro-inflammatory mediator that can activate leukocytes, platelets, and endothelial cells, all of which play a role in vaso-occlusion. We hypothesize that hypoxia-reoxygenation (H/R) in SCD mice activates complement, promoting inflammation and vaso-occlusion. At baseline and after H/R, sickle Townes-SS mice had increased C3 activation fragments and C5b-9 deposition in kidneys, livers and lungs and alternative pathway Bb fragments in plasma compared to control AA-mice. Activated complement promoted vaso-occlusion (microvascular stasis) in SS-mice; infusion of zymosan-activated, but not heat-inactivated serum, induced substantial vaso-occlusion in the skin venules of SS-mice. Infusion of recombinant C5a induced stasis in SS, but not AA-mice that was blocked by anti-C5a receptor (C5aR) IgG. C5a-mediated stasis was accompanied by inflammatory responses in SS-mice including NF-κB activation and increased expression of TLR4 and adhesion molecules VCAM-1, ICAM-1, and E-selectin in the liver. Anti-C5aR IgG blocked these inflammatory responses. Also, C5a rapidly up-regulated Weibel-Palade body P-selectin and von Willebrand factor on the surface of human umbilical vein endothelial cells in vitro and on vascular endothelium in vivo. In SS-mice, a blocking antibody to P-selectin inhibited C5a-induced stasis. Similarly, an antibody to C5 that blocks murine C5 cleavage or an antibody that blocks C5aR inhibited H/R-induced stasis in SS-mice. These results suggest that inhibition of C5a may be beneficial in SCD.


Asunto(s)
Anemia de Células Falciformes/inmunología , Anticuerpos Neutralizantes/farmacología , Trastornos Cerebrovasculares/inmunología , Complemento C3/inmunología , Complemento C5a/inmunología , Receptor de Anafilatoxina C5a/inmunología , Anemia de Células Falciformes/tratamiento farmacológico , Anemia de Células Falciformes/genética , Anemia de Células Falciformes/patología , Animales , Trastornos Cerebrovasculares/tratamiento farmacológico , Trastornos Cerebrovasculares/genética , Trastornos Cerebrovasculares/patología , Complemento C3/genética , Complemento C5a/antagonistas & inhibidores , Complemento C5a/genética , Complejo de Ataque a Membrana del Sistema Complemento/genética , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Modelos Animales de Enfermedad , Selectina E/genética , Selectina E/inmunología , Regulación de la Expresión Génica , Humanos , Inmunidad Innata , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/inmunología , Riñón/irrigación sanguínea , Riñón/efectos de los fármacos , Riñón/inmunología , Riñón/patología , Hígado/irrigación sanguínea , Hígado/efectos de los fármacos , Hígado/inmunología , Hígado/patología , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Masculino , Ratones , Ratones Transgénicos , FN-kappa B/genética , FN-kappa B/inmunología , Selectina-P/antagonistas & inhibidores , Selectina-P/genética , Selectina-P/inmunología , Receptor de Anafilatoxina C5a/antagonistas & inhibidores , Receptor de Anafilatoxina C5a/genética , Transducción de Señal , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/inmunología
7.
J Immunol ; 197(5): 1762-75, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27474078

RESUMEN

Recently, we demonstrated that IgG Abs can organize into ordered hexamers after binding their cognate Ags expressed on cell surfaces. This process is dependent on Fc:Fc interactions, which promote C1q binding, the first step in classical pathway complement activation. We went on to engineer point mutations that stimulated IgG hexamer formation and complement-dependent cytotoxicity (CDC). The hexamer formation-enhanced (HexaBody) CD20 and CD38 mAbs support faster, more robust CDC than their wild-type counterparts. To further investigate the CDC potential of these mAbs, we used flow cytometry, high-resolution digital imaging, and four-color confocal microscopy to examine their activity against B cell lines and primary chronic lymphocytic leukemia cells in sera depleted of single complement components. We also examined the CDC activity of alemtuzumab (anti-CD52) and mAb W6/32 (anti-HLA), which bind at high density to cells and promote substantial complement activation. Although we observed little CDC for mAb-opsonized cells reacted with sera depleted of early complement components, we were surprised to discover that the Hexabody mAbs, as well as ALM and W6/32, were all quite effective at promoting CDC in sera depleted of individual complement components C6 to C9. However, neutralization studies conducted with an anti-C9 mAb verified that C9 is required for CDC activity against cell lines. These highly effective complement-activating mAbs efficiently focus activated complement components on the cell, including C3b and C9, and promote CDC with a very low threshold of MAC binding, thus providing additional insight into their enhanced efficacy in promoting CDC.


Asunto(s)
ADP-Ribosil Ciclasa 1/metabolismo , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos CD20/metabolismo , Antígenos/inmunología , Sitios de Unión de Anticuerpos , Complemento C9/metabolismo , Glicoproteínas de Membrana/metabolismo , ADP-Ribosil Ciclasa 1/inmunología , Alemtuzumab , Anticuerpos Monoclonales Humanizados/inmunología , Antígenos CD20/inmunología , Linfocitos B/inmunología , Línea Celular Tumoral , Activación de Complemento , Complemento C3b/metabolismo , Complemento C9/inmunología , Proteínas del Sistema Complemento/inmunología , Proteínas del Sistema Complemento/metabolismo , Humanos , Glicoproteínas de Membrana/inmunología
8.
Clin Immunol ; 181: 24-28, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28578024

RESUMEN

We examined complement-dependent cytotoxicity (CDC) by hexamer formation-enhanced CD20 mAb Hx-7D8 of patient-derived chronic lymphocytic leukemia (CLL) cells that are relatively resistant to CDC. CDC was analyzed in normal human serum (NHS) and serum from an individual genetically deficient for C9. Hx-7D8 was able to kill up to 80% of CLL cells in complete absence of C9. We conclude that the narrow C5b-8 pores formed without C9 are sufficient for CDC due to efficient antibody-mediated hexamer formation. In the absence of C9, we observed transient intracellular increases of Ca2+ during CDC (as assessed with FLUO-4) that were extended in time. This suggests that small C5b-8 pores allow Ca2+ to enter the cell, while dissipation of the fluorescent signal accompanying cell disintegration is delayed. The Ca2+ signal is retained concomitantly with TOPRO-3 (viability dye) staining, thereby confirming that Ca2+ influx represents the most proximate mediator of cell death by CDC.


Asunto(s)
Complemento C9/deficiencia , Proteínas del Sistema Complemento/inmunología , Síndromes de Inmunodeficiencia/inmunología , Leucemia Linfocítica Crónica de Células B , Rituximab/farmacología , Calcio/metabolismo , Supervivencia Celular/efectos de los fármacos , Complemento C9/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/inmunología , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Proteínas del Sistema Complemento/metabolismo , Enfermedades por Deficiencia de Complemento Hereditario , Humanos , Inmunoterapia , Polimerizacion
9.
Blood ; 125(5): 762-6, 2015 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-25498911

RESUMEN

A specialized form of trogocytosis occurs when Fcγ receptors on acceptor cells take up and internalize donor cell-associated immune complexes composed of specific monoclonal antibodies (mAbs) bound to target antigens on donor cells. This trogocytosis reaction, an example of antigenic modulation, has been described in recent clinical correlative studies and in vitro investigations for several mAbs used in cancer immunotherapy, including rituximab and ofatumumab. We discuss the impact of Fcγ-receptor-mediated trogocytosis on the efficacy of cancer immunotherapy and other mAb-based therapies.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/farmacología , Anticuerpos Monoclonales/farmacología , Antígenos CD20/inmunología , Células Dendríticas/efectos de los fármacos , Subgrupos Linfocitarios/efectos de los fármacos , Receptores de IgG/inmunología , Anticuerpos Monoclonales Humanizados , Presentación de Antígeno , Complejo Antígeno-Anticuerpo/metabolismo , Antígenos CD20/genética , Antineoplásicos/farmacología , Células Dendríticas/citología , Células Dendríticas/inmunología , Expresión Génica , Historia del Siglo XXI , Humanos , Sinapsis Inmunológicas/efectos de los fármacos , Sinapsis Inmunológicas/inmunología , Inmunoterapia/historia , Células Asesinas Naturales/citología , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Subgrupos Linfocitarios/citología , Subgrupos Linfocitarios/inmunología , Monocitos/citología , Monocitos/efectos de los fármacos , Monocitos/inmunología , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Transporte de Proteínas , Receptores de IgG/genética , Rituximab
10.
Clin Immunol ; 171: 32-35, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27546448

RESUMEN

During malarial anemia, 20 uninfected red blood cells (RBCs) are destroyed for every RBC infected by Plasmodium falciparum (Pf). Increasing evidence indicates an important role for complement in destruction of uninfected RBCs. Products of RBC lysis induced by Pf, including the digestive vacuole and hematin, activate complement and promote C3 fragment deposition on uninfected RBCs. C3-opsonized cells are then subject to extravascular destruction mediated by fixed tissue macrophages which express receptors for C3 fragments. The Compstatin family of cyclic peptides blocks complement activation at the C3 cleavage step, and is under investigation for treatment of complement-mediated diseases. We demonstrate, that under a variety of stringent conditions, second-generation Compstatin analogue Cp40 completely blocks hematin-mediated deposition of C3 fragments on naïve RBCs. Our findings indicate that prophylactic provision of Compstatin for malaria-infected individuals at increased risk for anemia may provide a safe and inexpensive treatment to prevent or substantially reduce malarial anemia.


Asunto(s)
Complemento C3b/metabolismo , Eritrocitos/efectos de los fármacos , Hemina/metabolismo , Péptidos Cíclicos/farmacología , Anemia/tratamiento farmacológico , Eritrocitos/metabolismo , Humanos , Malaria Falciparum/tratamiento farmacológico , Plasmodium falciparum
11.
J Immunol ; 192(4): 1620-9, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24431228

RESUMEN

Ofatumumab (OFA), a human CD20-targeting mAb, kills B lymphocytes using the innate immune system including complement-dependent cytotoxicity (CDC). The efficacy of OFA in patients with chronic lymphocytic leukemia (CLL) is limited by drug resistance, which is not well characterized. To better understand mechanisms of resistance, we prospectively studied CLL cells isolated from blood samples collected before and after in vivo exposure to the initial dose of OFA therapy in 25 patients undergoing their first treatment for progressive CLL. As previously reported, OFA therapy rapidly decreased the absolute lymphocyte count, CD20 expression by CLL cells, and serum complement levels. We now show that after administration of the first dose of OFA, there was a modest rebound in the absolute lymphocyte count and serum complement levels, but substantial ongoing loss of CD20 expression by CLL cells. These post-OFA treatment CLL cells were highly resistant to OFA-mediated CDC but retained sensitivity to alemtuzumab-mediated CDC in vitro. Posttherapy serum OFA levels correlated inversely with both the amount of pretreatment circulating cell-bound CD20 and with the decrease in this value following treatment. In vitro OFA-mediated CDC did not predict clinical responses, and the patients with first-dose reactions to OFA did not have markers of increased complement activation in vivo. We propose that optimal efficacy of CD20- targeted therapy for CLL requires determining an mAb dose size and frequency that optimizes CLL killing without exceeding the capacity of the cytotoxic mechanisms and thus minimizes loss of CD20 expression in the surviving CLL cells.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antígenos CD20/inmunología , Linfocitos B/inmunología , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Adulto , Anciano , Anciano de 80 o más Años , Alemtuzumab , Anticuerpos Monoclonales/sangre , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antígenos CD/biosíntesis , Antígenos CD20/biosíntesis , Antígenos CD20/sangre , Antígenos de Neoplasias/biosíntesis , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Linfocitos B/citología , Linfocitos B/efectos de los fármacos , Antígeno CD52 , Proteínas del Sistema Complemento/metabolismo , Ciclofosfamida/uso terapéutico , Citotoxicidad Inmunológica/inmunología , Resistencia a Antineoplásicos , Femenino , Glicoproteínas/biosíntesis , Humanos , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Pentostatina/uso terapéutico , Resultado del Tratamiento
12.
J Immunol ; 192(10): 4844-51, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24729617

RESUMEN

Dysregulation of the complement alternative pathway can cause disease in various organs that may be life-threatening. Severe alternative pathway dysregulation can be triggered by autoantibodies to the C3 convertase, termed nephritic factors, which cause pathological stabilization of the convertase enzyme and confer resistance to innate control mechanisms; unregulated complement consumption followed by deposition of C3 fragments in tissues ensues. The mAb, 3E7, and its humanized derivative, H17, have been shown previously to specifically bind activated C3 and prevent binding of both the activating protein, factor B, and the inhibitor, factor H, which are opposite effects that complicate its potential for therapy. Using ligand binding assays, functional assays, and electron microscopy, we show that these Abs bind C3b via a site that overlaps the binding site on C3 for the Ba domain within factor B, thereby blocking an interaction essential for convertase formation. Both Abs also bind the preformed convertase, C3bBb, and provide powerful inhibition of complement activation by preventing cleavage of C3. Critically, the Abs also bound and inhibited C3 cleavage by the nephritic factor-stabilized convertase. We suggest that by preventing enzyme formation and/or cleavage of C3 to its active downstream fragments, H17 may be an effective therapy for conditions caused by severe dysregulation of the C3 convertase and, in particular, those that involve nephritic factors, such as dense deposit disease.


Asunto(s)
Anticuerpos Monoclonales Humanizados/inmunología , C3 Convertasa de la Vía Alternativa del Complemento/inmunología , Convertasas de Complemento C3-C5/inmunología , Factor B del Complemento/inmunología , Vía Alternativa del Complemento/inmunología , Enfermedades Renales/inmunología , Anticuerpos Monoclonales Humanizados/uso terapéutico , C3 Convertasa de la Vía Alternativa del Complemento/antagonistas & inhibidores , Vía Alternativa del Complemento/efectos de los fármacos , Humanos , Enfermedades Renales/tratamiento farmacológico , Enfermedades Renales/patología
13.
Methods ; 65(1): 18-27, 2014 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-23886909

RESUMEN

The ability of complement to promote lysis of antibody-opsonized cells is well-established. Virtually all of the molecular details of this reaction have been elucidated and numerous points of regulation have also been delineated. Use of this information, along with the techniques that were first applied in the fundamental studies of complement, has allowed for investigations of the role of complement in mAb-based immunotherapies of cancer. These studies, which have often combined in vitro investigations with parallel correlative clinical measurements, have revealed that several FDA-approved mAbs make use of complement as an effector function in promoting opsonization and killing of targeted malignant cells. We describe the key methods used in this work, and discuss how the results of these studies provide rational approaches for making more effective use of complement in mAb-based cancer immunotherapy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Activación de Complemento/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Animales , Anticuerpos Monoclonales/farmacología , Antineoplásicos/farmacología , Proteínas del Sistema Complemento/fisiología , Citotoxicidad Inmunológica , Ensayos de Selección de Medicamentos Antitumorales , Humanos , Neoplasias/inmunología
14.
Mol Pharmacol ; 86(5): 485-91, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24944188

RESUMEN

Since approval of rituximab for treatment of B cell non-Hodgkin lymphoma, development of monoclonal antibodies (mAbs) for cancer treatment and elucidation of their cytotoxic mechanisms have been subject to intense investigations. Compelling evidence indicates that rituximab and another CD20 mAb, ofatumumab, must use the body's cellular and humoral immune effector functions to kill malignant cells. Other U.S. Food and Drug Administration-approved mAbs, including obinutuzumab, cetuximab, and trastuzumab, require, in part, these effector mechanisms to eliminate tumor cells. Although gram quantities of mAbs can be administered to patients, our investigations of CD20 mAb-based therapies for chronic lymphocytic leukemia (CLL), including correlative measurements in clinical trials and studies with primary cells and cell lines, indicate that effector mechanisms necessary for mAb activity can be saturated or exhausted if tumor burdens are high, thus substantially compromising the efficacy of high-dose mAb therapy. Under these conditions, another reaction (trogocytosis) predominates in which bound CD20 mAb and CD20 are removed from targeted cells by effector cells that express Fcγ receptors, thereby allowing malignant cells to escape unharmed and continue to promote disease pathology. To address this problem, we propose that a low-dose strategy, based on administering 30-50 mg of CD20 mAb three times per week, may be far more effective for CLL than standard dosing because it will minimize effector function saturation and reduce trogocytosis. This approach may have general applicability to other mAbs that use immune effector functions, and could be formulated into a subcutaneous treatment strategy that would be more accessible and possibly more efficacious for patients.


Asunto(s)
Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Antígenos CD20/inmunología , Antineoplásicos/administración & dosificación , Antineoplásicos/inmunología , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Animales , Ensayos Clínicos Fase III como Asunto , Humanos
15.
Blood ; 119(26): 6307-16, 2012 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-22577173

RESUMEN

Paroxysmal nocturnal hemoglobinuria (PNH) is characterized by complement-mediated intravascular hemolysis because of the lack from erythrocyte surface of the complement regulators CD55 and CD59, with subsequent uncontrolled continuous spontaneous activation of the complement alternative pathway (CAP), and at times of the complement classic pathway. Here we investigate in an in vitro model the effect on PNH erythrocytes of a novel therapeutic strategy for membrane-targeted delivery of a CAP inhibitor. TT30 is a 65 kDa recombinant human fusion protein consisting of the iC3b/C3d-binding region of complement receptor 2 (CR2) and the inhibitory domain of the CAP regulator factor H (fH). TT30 completely inhibits in a dose-dependent manner hemolysis of PNH erythrocytes in a modified extended acidified serum assay, and also prevents C3 fragment deposition on surviving PNH erythrocytes. The efficacy of TT30 derives from its direct binding to PNH erythrocytes; if binding to the erythrocytes is disrupted, only partial inhibition of hemolysis is mediated by TT30 in solution, which is similar to that produced by the fH moiety of TT30 alone, or by intact human fH. TT30 is a membrane-targeted selective CAP inhibitor that may prevent both intravascular and C3-mediated extravascular hemolysis of PNH erythrocytes and warrants consideration for the treatment of PNH patients.


Asunto(s)
Factor H de Complemento/química , Eritrocitos/efectos de los fármacos , Hemoglobinuria Paroxística/sangre , Hemólisis/efectos de los fármacos , Proteínas de Fusión Oncogénica/farmacología , Receptores de Complemento 3d/química , Proteínas Recombinantes de Fusión/farmacología , Estudios de Casos y Controles , Células Cultivadas , Complemento C3/efectos adversos , Complemento C3/antagonistas & inhibidores , Complemento C3/farmacología , Factor H de Complemento/metabolismo , Factor H de Complemento/farmacología , Proteínas del Sistema Complemento/efectos adversos , Proteínas del Sistema Complemento/fisiología , Citoprotección/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Eritrocitos/fisiología , Hemoglobinuria Paroxística/patología , Humanos , Proteínas de Fusión Oncogénica/metabolismo , Unión Proteica , Receptores de Complemento 3d/metabolismo , Receptores de Complemento 3d/fisiología , Proteínas Recombinantes de Fusión/metabolismo
16.
Am J Hematol ; 89(7): 757-65, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24723493

RESUMEN

Chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL) patients with purine analog refractory disease or TP53 dysfunction still have limited treatment options and poor survival. Alemtuzumab-containing chemoimmunotherapy regimens can be effective but frequently cause serious infections. We report a Phase II trial testing the efficacy and tolerability of a short-duration regimen combining pentostatin, alemtuzumab, and low-dose high-frequency rituximab designed to decrease the risk of treatment-associated infections and to limit the loss of CD20 expression by CLL cells. The study enrolled 39 patients with progressive CLL that was either relapsed/refractory (n = 36) or previously untreated with 17p13 deletion (17p13-) (n = 3). Thirteen (33%) patients had both 17p13- and TP53 mutations predicted to be dysfunctional, and eight patients had purine analog refractory CLL without TP53 dysfunction. Twenty-six (67%) patients completed therapy, with only five (13%) patients having treatment-limiting toxicity and no treatment-related deaths. Twenty-two (56%) patients responded to treatment, with 11 (28%) complete responses (four with incomplete bone marrow recovery). Median progression-free survival was 7.2 months, time to next treatment was 9.1 months, and overall survival was 34.1 months. The majority of deaths (82%) were caused by progressive disease, including transformed diffuse large B-cell lymphoma (n = 6). Correlative studies showed that low-dose rituximab activates complement and natural killer cells without a profound and sustained decrease in expression of CD20 by circulating CLL cells. We conclude that pentostatin, alemtuzumab, and low-dose high-frequency rituximab is a tolerable and effective therapy for CLL and that low-dose rituximab therapy can activate innate immune cytotoxic mechanisms without substantially decreasing CD20 expression.


Asunto(s)
Antígenos CD20/biosíntesis , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Anciano , Alemtuzumab , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales de Origen Murino/administración & dosificación , Anticuerpos Monoclonales de Origen Murino/efectos adversos , Antígenos CD20/inmunología , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Deleción Cromosómica , Cromosomas Humanos Par 17 , Supervivencia sin Enfermedad , Femenino , Humanos , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/inmunología , Masculino , Persona de Mediana Edad , Células Neoplásicas Circulantes/inmunología , Pentostatina/administración & dosificación , Pentostatina/efectos adversos , Recurrencia , Inducción de Remisión , Rituximab
17.
J Immunol ; 188(7): 3532-41, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22368276

RESUMEN

The CD20 mAb ofatumumab (OFA) induces complement-mediated lysis of B cells. In an investigator-initiated phase II trial of OFA plus chemotherapy for chronic lymphocytic leukemia (CLL), OFA treatment promoted partial CLL B cell depletion that coincided with reduced complement titers. Remaining CLL B cells circulated with bound OFA and covalently bound complement breakdown product C3d, indicative of ongoing complement activation. Presumably, neither complement- nor effector cell-based mechanisms were sufficiently robust to clear these remaining B cells. Instead, almost all of the bound OFA and CD20 was removed from the cells, in accordance with previous clinical studies that demonstrated comparable loss of CD20 from B cells after treatment of CLL patients with rituximab. In vitro experiments with OFA and rituximab addressing these observations suggest that host effector mechanisms that support mAb-mediated lysis and tumor cell clearance are finite, and they can be saturated or exhausted at high B cell burdens, particularly at high mAb concentrations. Interestingly, only a fraction of available complement was required to kill cells with CD20 mAbs, and killing could be tuned by titrating the mAb concentration. Consequently, maximal B cell killing of an initial and secondary B cell challenge was achieved with intermediate mAb concentrations, whereas high concentrations promoted lower overall killing. Therefore, mAb therapies that rely substantially on effector mechanisms subject to exhaustion, including complement, may benefit from lower, more frequent dosing schemes optimized to sustain and maximize killing by cytotoxic immune effector systems.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales de Origen Murino/uso terapéutico , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Proteínas del Sistema Complemento/inmunología , Inmunoterapia , Leucemia Linfocítica Crónica de Células B/terapia , Alemtuzumab , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales de Origen Murino/administración & dosificación , Citotoxicidad Celular Dependiente de Anticuerpos , Antígenos CD20/inmunología , Antígenos de Neoplasias/inmunología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Subgrupos de Linfocitos B/efectos de los fármacos , Subgrupos de Linfocitos B/inmunología , Línea Celular Tumoral/efectos de los fármacos , Terapia Combinada , Activación de Complemento , Complemento C3b/análisis , Complemento C4b/análisis , Ciclofosfamida/administración & dosificación , Citotoxicidad Inmunológica , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/inmunología , Leucemia Linfocítica Crónica de Células B/patología , Recuento de Linfocitos , Proteínas Opsoninas/inmunología , Rituximab , Vidarabina/administración & dosificación , Vidarabina/análogos & derivados
18.
Haematologica ; 98(8): 1259-63, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23716541

RESUMEN

Rituximab is an effective treatment for autoimmune cytopenias associated with chronic lymphocytic leukemia. Despite the incorporation of rituximab into fludarabine-based chemotherapy regimens, the incidence of autoimmune cytopenias has remained high. Inadequate rituximab exposure due to rapid antibody clearance may be a contributing factor. To test this hypothesis, we measured serum rituximab levels in patients treated with fludarabine and rituximab (375 mg/m(2)). All patients had undetectable rituximab trough levels by the end of cycle 1, and one-third had undetectable levels already on Day 6 of cycle 1. Although rituximab trough levels increased progressively with each cycle, only by cycle 4 did the median trough level exceed 10 ug/mL. The median half-life of rituximab during cycle 1 was 27 hours, compared to 199 hours during cycle 4 (P<0.0001). There was a significant inverse correlation between the rituximab half-life in cycle 1 and the degree of tumor burden (P=0.02). Two patients who were identified as having subclinical autoimmune hemolysis prior to therapy were given additional doses of rituximab during the initial cycles of therapy and did not develop clinically significant hemolysis. One patient who developed clinically significant hemolysis during therapy was given additional rituximab doses during cycles 3-5 and was able to successfully complete his treatment. In conclusion, rituximab is cleared so rapidly during the initial cycles of therapy for chronic lymphocytic leukemia that most patients have only transient serum levels. More frequent dosing of rituximab may be required to prevent autoimmune complications in at-risk patients (clinicaltrials.gov identifier:00001586).


Asunto(s)
Anemia Hemolítica Autoinmune/sangre , Anemia Hemolítica Autoinmune/inducido químicamente , Anticuerpos Monoclonales de Origen Murino/sangre , Antineoplásicos/sangre , Leucemia Linfocítica Crónica de Células B/sangre , Tasa de Depuración Metabólica/fisiología , Adulto , Anciano , Anticuerpos Monoclonales de Origen Murino/administración & dosificación , Anticuerpos Monoclonales de Origen Murino/efectos adversos , Antineoplásicos/administración & dosificación , Estudios de Cohortes , Femenino , Humanos , Inmunoterapia/efectos adversos , Incidencia , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Masculino , Tasa de Depuración Metabólica/efectos de los fármacos , Persona de Mediana Edad , Rituximab , Factores de Tiempo , Resultado del Tratamiento , Vidarabina/administración & dosificación , Vidarabina/efectos adversos , Vidarabina/análogos & derivados , Vidarabina/sangre
19.
J Immunol ; 187(6): 3438-47, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21841127

RESUMEN

We previously reported that 1 h after infusion of CD20 mAb rituximab in patients with chronic lymphocytic leukemia (CLL), >80% of CD20 was removed from circulating B cells, and we replicated this finding, based on in vitro models. This reaction occurs via an endocytic process called shaving/trogocytosis, mediated by FcγR on acceptor cells including monocytes/macrophages, which remove and internalize rituximab-CD20 immune complexes from B cells. Beers et al. reported that CD20 mAb-induced antigenic modulation occurs as a result of internalization of B cell-bound mAb-CD20 complexes by the B cells themselves, with internalization of ∼40% observed after 2 h at 37°C. These findings raise fundamental questions regarding the relative importance of shaving versus internalization in promoting CD20 loss and have substantial implications for the design of mAb-based cancer therapies. Therefore, we performed direct comparisons, based on flow cytometry, to determine the relative rates and extent of shaving versus internalization. B cells, from cell lines, from patients with CLL, and from normal donors, were opsonized with CD20 mAbs rituximab or ofatumumab and incubated for varying times and then reacted with acceptor THP-1 monocytes to promote shaving. We find that shaving induces considerably greater loss of CD20 and bound mAb from opsonized B cells in much shorter time periods (75-90% in <45 min) than is observed for internalization. Both shaving/trogocytosis and internalization could contribute to CD20 loss when CLL patients receive rituximab therapy, but shaving should occur more rapidly and is most likely to be the key mechanism of CD20 loss.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/farmacología , Antígenos CD20/metabolismo , Antineoplásicos/farmacología , Linfocitos B/metabolismo , Endocitosis/fisiología , Monocitos/metabolismo , Animales , Linfocitos B/efectos de los fármacos , Línea Celular , Separación Celular , Endocitosis/efectos de los fármacos , Citometría de Flujo , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/metabolismo , Ratones , Receptores de IgG/metabolismo , Rituximab , Transfección
20.
Curr Protoc ; 3(10): e897, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37830752

RESUMEN

Trogocytosis is a process in which receptors on acceptor cells remove and internalize cognate ligands from donor cells. Trogocytosis has a profound and negative impact on mAb-based cancer immunotherapy, as seen in the treatment of chronic lymphocytic leukemia (CLL) with CD20 mAbs, such as rituximab (RTX) and ofatumumab (OFA). Our clinical observations of RTX/OFA-mediated loss of the CD20 target from circulating CLL cells have been replicated in our in vitro studies. Here we describe flow cytometry and fluorescence microscopy experiments, which demonstrate that acceptor cells, such as monocytes/macrophages that express FcγR, remove and internalize both antigen and donor cell-bound cognate IgG mAbs for several different mAb-donor cell pairs. Fluorescent mAbs and portions of the plasma cell membrane are transferred from donor cells to acceptor cells, which include the THP-1 monocytic cell line as well as freshly isolated monocytes. We describe rigorous controls to validate the reactions and eliminate dissociation or internalization as alternative mechanisms. Trogocytosis is likely to contribute to neutropenia, thrombocytopenia, and liver damage associated with use of antibody-drug conjugates. The methods we have described should allow for examination of strategies focused on blocking trogocytosis and its adverse effects. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Trogocytosis of mAb-opsonized donor cells mediated by adherent THP-1 cells Alternate Protocol: Application of fluorescence microscopy to examine THP-1 cell-mediated trogocytosis Support Protocol 1: Alexa labeling of mAbs and determination of F/P ratios Support Protocol 2: Standard washing procedure Support Protocol 3: Labeling and opsonization of cells Basic Protocol 2: Trogocytosis mediated by human monocytes as acceptor cells Support Protocol 4: Isolation of human monocytes Basic Protocol 3: Trogocytosis mediated by THP-1 cells in solution Support Protocol 5: Retinoic acid treatment of THP-1 cells Support Protocol 6: Culturing of SCC-25, BT-474, MOLT-4 and THP-1 cell lines.


Asunto(s)
Leucemia Linfocítica Crónica de Células B , Humanos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/metabolismo , Anticuerpos Monoclonales de Origen Murino/uso terapéutico , Trogocitosis , Antígenos CD20/uso terapéutico , Rituximab/farmacología , Rituximab/uso terapéutico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA