Cargando…

A Fully Human IgE Specific for CD38 as a Potential Therapy for Multiple Myeloma

SIMPLE SUMMARY: Multiple myeloma (MM) is blood cancer of plasma cells. Plasma cells are white blood cells, which are part of the immune system. Malignant plasma cells are found in the bone marrow and are difficult to treat. There have been many new therapies developed in recent years, but the diseas...

Descripción completa

Detalles Bibliográficos
Autores principales: Candelaria, Pierre V., Nava, Miguel, Daniels-Wells, Tracy R., Penichet, Manuel L.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10526502/
https://www.ncbi.nlm.nih.gov/pubmed/37760502
http://dx.doi.org/10.3390/cancers15184533
_version_ 1785111037388783616
author Candelaria, Pierre V.
Nava, Miguel
Daniels-Wells, Tracy R.
Penichet, Manuel L.
author_facet Candelaria, Pierre V.
Nava, Miguel
Daniels-Wells, Tracy R.
Penichet, Manuel L.
author_sort Candelaria, Pierre V.
collection PubMed
description SIMPLE SUMMARY: Multiple myeloma (MM) is blood cancer of plasma cells. Plasma cells are white blood cells, which are part of the immune system. Malignant plasma cells are found in the bone marrow and are difficult to treat. There have been many new therapies developed in recent years, but the disease is still considered incurable. Some of the treatments for MM are antibodies of the IgG class. We have developed a fully human antibody of another antibody class, IgE, targeting the CD38 molecule that is expressed on the surface of several cancers including MM. Our goal is to use the anti-CD38 IgE antibody to recruit different types of immune cells to kill malignant cells. In this article, we report that the antibody shows anti-cancer effects against MM cells. Thus, this IgE antibody should be further explored as a novel treatment for MM. ABSTRACT: 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.
format Online
Article
Text
id pubmed-10526502
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-105265022023-09-28 A Fully Human IgE Specific for CD38 as a Potential Therapy for Multiple Myeloma Candelaria, Pierre V. Nava, Miguel Daniels-Wells, Tracy R. Penichet, Manuel L. Cancers (Basel) Article SIMPLE SUMMARY: Multiple myeloma (MM) is blood cancer of plasma cells. Plasma cells are white blood cells, which are part of the immune system. Malignant plasma cells are found in the bone marrow and are difficult to treat. There have been many new therapies developed in recent years, but the disease is still considered incurable. Some of the treatments for MM are antibodies of the IgG class. We have developed a fully human antibody of another antibody class, IgE, targeting the CD38 molecule that is expressed on the surface of several cancers including MM. Our goal is to use the anti-CD38 IgE antibody to recruit different types of immune cells to kill malignant cells. In this article, we report that the antibody shows anti-cancer effects against MM cells. Thus, this IgE antibody should be further explored as a novel treatment for MM. ABSTRACT: 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. MDPI 2023-09-13 /pmc/articles/PMC10526502/ /pubmed/37760502 http://dx.doi.org/10.3390/cancers15184533 Text en © 2023 by the authors. https://creativecommons.org/licenses/by/4.0/Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Candelaria, Pierre V.
Nava, Miguel
Daniels-Wells, Tracy R.
Penichet, Manuel L.
A Fully Human IgE Specific for CD38 as a Potential Therapy for Multiple Myeloma
title A Fully Human IgE Specific for CD38 as a Potential Therapy for Multiple Myeloma
title_full A Fully Human IgE Specific for CD38 as a Potential Therapy for Multiple Myeloma
title_fullStr A Fully Human IgE Specific for CD38 as a Potential Therapy for Multiple Myeloma
title_full_unstemmed A Fully Human IgE Specific for CD38 as a Potential Therapy for Multiple Myeloma
title_short A Fully Human IgE Specific for CD38 as a Potential Therapy for Multiple Myeloma
title_sort fully human ige specific for cd38 as a potential therapy for multiple myeloma
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10526502/
https://www.ncbi.nlm.nih.gov/pubmed/37760502
http://dx.doi.org/10.3390/cancers15184533
work_keys_str_mv AT candelariapierrev afullyhumanigespecificforcd38asapotentialtherapyformultiplemyeloma
AT navamiguel afullyhumanigespecificforcd38asapotentialtherapyformultiplemyeloma
AT danielswellstracyr afullyhumanigespecificforcd38asapotentialtherapyformultiplemyeloma
AT penichetmanuell afullyhumanigespecificforcd38asapotentialtherapyformultiplemyeloma
AT candelariapierrev fullyhumanigespecificforcd38asapotentialtherapyformultiplemyeloma
AT navamiguel fullyhumanigespecificforcd38asapotentialtherapyformultiplemyeloma
AT danielswellstracyr fullyhumanigespecificforcd38asapotentialtherapyformultiplemyeloma
AT penichetmanuell fullyhumanigespecificforcd38asapotentialtherapyformultiplemyeloma