12 results on '"Haag, Friedrich"'
Search Results
2. Nanobody-based CD38-specific heavy chain antibodies induce killing of multiple myeloma and other hematological malignancies.
- Author
-
Schriewer L, Schütze K, Petry K, Hambach J, Fumey W, Koenigsdorf J, Baum N, Menzel S, Rissiek B, Riecken K, Fehse B, Röckendorf JL, Schmid J, Albrecht B, Pinnschmidt H, Ayuk F, Kröger N, Binder M, Schuch G, Hansen T, Haag F, Adam G, Koch-Nolte F, and Bannas P
- Subjects
- Aged, Animals, Cell Line, Tumor, Epitopes immunology, Female, Humans, Male, Mice, Mice, SCID, Middle Aged, ADP-ribosyl Cyclase 1 immunology, Antibodies, Monoclonal, Humanized therapeutic use, Hematologic Neoplasms drug therapy, Immunoglobulin G therapeutic use, Immunoglobulin Heavy Chains therapeutic use, Membrane Glycoproteins immunology, Multiple Myeloma drug therapy, Single-Domain Antibodies therapeutic use
- Abstract
Rationale : CD38 is a target for the therapy of multiple myeloma (MM) with monoclonal antibodies such as daratumumab and isatuximab. Since MM patients exhibit a high rate of relapse, the development of new biologics targeting alternative CD38 epitopes is desirable. The discovery of single-domain antibodies (nanobodies) has opened the way for a new generation of antitumor therapeutics. We report the generation of nanobody-based humanized IgG1 heavy chain antibodies (hcAbs) with a high specificity and affinity that recognize three different and non-overlapping epitopes of CD38 and compare their cytotoxicity against CD38-expressing hematological cancer cells in vitro , ex vivo and in vivo . Methods : We generated three humanized hcAbs (WF211-hcAb, MU1067-hcAb, JK36-hcAb) that recognize three different non-overlapping epitopes (E1, E2, E3) of CD38 by fusion of llama-derived nanobodies to the hinge- and Fc-domains of human IgG1. WF211-hcAb shares the binding epitope E1 with daratumumab. We compared the capacity of these CD38-specific hcAbs and daratumumab to induce CDC and ADCC in CD38-expressing tumor cell lines in vitro and in patient MM cells ex vivo as well as effects on xenograft tumor growth and survival in vivo . Results : CD38-specific heavy chain antibodies (WF211-hcAb, MU1067-hcAb, JK36-hcAb) potently induced antibody-dependent cellular cytotoxicity (ADCC) in CD38-expressing tumor cell lines and in primary patient MM cells, but only little if any complement-dependent cytotoxicity (CDC). In vivo, CD38-specific heavy chain antibodies significantly reduced the growth of systemic lymphomas and prolonged survival of tumor bearing SCID mice. Conclusions : CD38-specific nanobody-based humanized IgG1 heavy chain antibodies mediate cytotoxicity against CD38-expressing hematological cancer cells in vitro , ex vivo and in vivo . These promising results of our study indicate that CD38-specific hcAbs warrant further clinical development as therapeutics for multiple myeloma and other hematological malignancies., Competing Interests: Competing Interests: K.S., W.F., L.S., S.M., P.B., and F.K.-N. are co-inventors on a patent application on CD38-specific nanobodies. F.H. and F.K.-N. receive a share of antibody and protein sales via MediGate GmbH, a wholly owned subsidiary of the University Medical Center Hamburg-Eppendorf., (© The author(s).)
- Published
- 2020
- Full Text
- View/download PDF
3. Targeting CD38-Expressing Multiple Myeloma and Burkitt Lymphoma Cells In Vitro with Nanobody-Based Chimeric Antigen Receptors (Nb-CARs).
- Author
-
Hambach J, Riecken K, Cichutek S, Schütze K, Albrecht B, Petry K, Röckendorf JL, Baum N, Kröger N, Hansen T, Schuch G, Haag F, Adam G, Fehse B, Bannas P, and Koch-Nolte F
- Subjects
- Bone Marrow pathology, Cell Line, Tumor, Cytotoxicity, Immunologic, Humans, Killer Cells, Natural metabolism, Lentivirus metabolism, Luciferases metabolism, Luminescence, Models, Molecular, ADP-ribosyl Cyclase 1 metabolism, Burkitt Lymphoma metabolism, Multiple Myeloma metabolism, Receptors, Chimeric Antigen metabolism, Single-Domain Antibodies metabolism
- Abstract
The NAD-hydrolyzing ecto-enzyme CD38 is overexpressed by multiple myeloma and other hematological malignancies. We recently generated CD38-specific nanobodies, single immunoglobulin variable domains derived from heavy-chain antibodies naturally occurring in llamas. Nanobodies exhibit high solubility and stability, allowing easy reformatting into recombinant fusion proteins. Here we explore the utility of CD38-specific nanobodies as ligands for nanobody-based chimeric antigen receptors (Nb-CARs). We cloned retroviral expression vectors for CD38-specific Nb-CARs. The human natural killer cell line NK-92 was transduced to stably express these Nb-CARs. As target cells we used CD38-expressing as well as CRISPR/Cas9-generated CD38-deficient tumor cell lines (CA-46, LP-1, and Daudi) transduced with firefly luciferase. With these effector and target cells we established luminescence and flow-cytometry CAR-dependent cellular cytotoxicity assays (CARDCCs). Finally, the cytotoxic efficacy of Nb-CAR NK-92 cells was tested on primary patient-derived CD38-expressing multiple myeloma cells. NK-92 cells expressing CD38-specific Nb-CARs specifically lysed CD38-expressing but not CD38-deficient tumor cell lines. Moreover, the Nb-CAR-NK cells effectively depleted CD38-expressing multiple myeloma cells in primary human bone marrow samples. Our results demonstrate efficacy of Nb-CARs in vitro. The potential clinical efficacy of Nb-CARs in vivo remains to be evaluated., Competing Interests: K.S., P.B., and F.K.-N. are co-inventors on a patent application on CD38-specific nanobodies. F.H. and F.K.-N. receive a share of antibody and protein sales via MediGate GmbH, a wholly owned subsidiary of the University Medical Center Hamburg-Eppendorf. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.
- Published
- 2020
- Full Text
- View/download PDF
4. CD38-Specific Biparatopic Heavy Chain Antibodies Display Potent Complement-Dependent Cytotoxicity Against Multiple Myeloma Cells.
- Author
-
Schütze K, Petry K, Hambach J, Schuster N, Fumey W, Schriewer L, Röckendorf J, Menzel S, Albrecht B, Haag F, Stortelers C, Bannas P, and Koch-Nolte F
- Subjects
- Animals, Antibodies, Bispecific genetics, Antibodies, Monoclonal pharmacology, Antibody-Dependent Cell Cytotoxicity, Camelids, New World, Cell Line, Tumor, Complement System Proteins metabolism, Humans, Immunoglobulin Heavy Chains genetics, Multiple Myeloma therapy, Recombinant Fusion Proteins genetics, Single-Domain Antibodies genetics, ADP-ribosyl Cyclase 1 immunology, Antibodies, Bispecific pharmacology, Antigens, Neoplasm immunology, Antineoplastic Agents pharmacology, Epitopes, B-Lymphocyte immunology, Immunotherapy methods, Multiple Myeloma immunology
- Abstract
CD38 is overexpressed by multiple myeloma cells and has emerged as a target for therapeutic antibodies. Nanobodies are soluble single domain antibody fragments derived from the VHH variable domain of heavy chain antibodies naturally occurring in camelids. We previously identified distinct llama nanobodies that recognize three non-overlapping epitopes of the extracellular domain of CD38. Here, we fused these VHH domains to the hinge, CH2, and CH3 domains of human IgG1, yielding highly soluble chimeric llama/human heavy chain antibodies (hcAbs). We analyzed the capacity of these hcAbs to mediate complement-dependent cytotoxicity (CDC) to CD38-expressing human multiple myeloma and Burkitt lymphoma cell lines. Combinations of two hcAbs that recognize distinct, non-overlapping epitopes of CD38 mediated potent CDC, in contrast to the hcAb monotherapy with only weak CDC capacity. Similarly, combining daratumumab with a hcAb that recognizes a non-overlapping epitope resulted in dramatically enhanced CDC. Further, introducing the E345R HexaBody mutation into the CH3 domain strongly enhanced the CDC potency of hcAbs to CD38-expressing cells. Exploiting their high solubility, we genetically fused two distinct nanobodies into heteromeric dimers via a flexible peptide linker and then fused these nanobody dimers to the hinge, CH2 and CH3 domains of human IgG1, yielding highly soluble, biparatopic hcAbs. These biparatopic hcAbs elicited CDC toward CD38-expressing myeloma cells more effectively than daratumumab. Our results underscore the advantage of nanobodies vs. pairs of VH and VL domains for constructing bispecific antibodies. Moreover, the CD38-specific biparatopic heavy chain antibodies described here represent potential new powerful therapeutics for treatment of multiple myeloma.
- Published
- 2018
- Full Text
- View/download PDF
5. Nanobodies effectively modulate the enzymatic activity of CD38 and allow specific imaging of CD38 + tumors in mouse models in vivo.
- Author
-
Fumey W, Koenigsdorf J, Kunick V, Menzel S, Schütze K, Unger M, Schriewer L, Haag F, Adam G, Oberle A, Binder M, Fliegert R, Guse A, Zhao YJ, Cheung Lee H, Malavasi F, Goldbaum F, van Hegelsom R, Stortelers C, Bannas P, and Koch-Nolte F
- Subjects
- ADP-ribosyl Cyclase 1 immunology, Animals, Camelids, New World, Cell Line, Tumor, Cell Surface Display Techniques, Disease Models, Animal, Epitopes immunology, Fluorescent Dyes, Humans, Membrane Glycoproteins immunology, Mice, Mice, Nude, Multiple Myeloma pathology, Xenograft Model Antitumor Assays, ADP-ribosyl Cyclase 1 metabolism, Antibodies, Monoclonal therapeutic use, Antineoplastic Agents therapeutic use, Membrane Glycoproteins metabolism, Multiple Myeloma diagnosis, Multiple Myeloma drug therapy, Single-Domain Antibodies immunology
- Abstract
The cell surface ecto-enzyme CD38 is a promising target antigen for the treatment of hematological malignancies, as illustrated by the recent approval of daratumumab for the treatment of multiple myeloma. Our aim was to evaluate the potential of CD38-specific nanobodies as novel diagnostics for hematological malignancies. We successfully identified 22 CD38-specific nanobody families using phage display technology from immunized llamas. Crossblockade analyses and in-tandem epitope binning revealed that the nanobodies recognize three different non-overlapping epitopes, with four nanobody families binding complementary to daratumumab. Three nanobody families inhibit the enzymatic activity of CD38 in vitro, while two others were found to act as enhancers. In vivo, fluorochrome-conjugated CD38 nanobodies efficiently reach CD38 expressing tumors in a rodent model within 2 hours after intravenous injection, thereby allowing for convenient same day in vivo tumor imaging. These nanobodies represent highly specific tools for modulating the enzymatic activity of CD38 and for diagnostic monitoring CD38-expressing tumors.
- Published
- 2017
- Full Text
- View/download PDF
6. A transplant "immunome" screening platform defines a targetable epitope fingerprint of multiple myeloma.
- Author
-
Schieferdecker A, Oberle A, Thiele B, Hofmann F, Göthel M, Miethe S, Hust M, Braig F, Voigt M, von Pein UM, Koch-Nolte F, Haag F, Alawi M, Indenbirken D, Grundhoff A, Bokemeyer C, Bacher U, Kröger N, and Binder M
- Subjects
- Adult, Aged, Cell Line, Tumor, Female, HL-60 Cells, Humans, Immunotherapy methods, Jurkat Cells, Male, Middle Aged, Multiple Myeloma immunology, Myeloma Proteins analysis, Myeloma Proteins isolation & purification, Peptide Library, Peptide Mapping methods, Proteome analysis, Epitope Mapping methods, Epitopes metabolism, Molecular Targeted Therapy methods, Multiple Myeloma therapy, Proteome immunology, Transplants immunology, Transplants metabolism
- Abstract
Multiple myeloma (MM) is a hematological cancer for which immune-based treatments are currently in development. Many of these rely on the identification of highly disease-specific, strongly and stably expressed antigens. Here, we profiled the myeloma B-cell immunome both to explore its predictive role in the context of autologous and allogeneic hematopoietic stem cell transplantation (HSCT) and to identify novel immunotherapeutic targets. We used random peptide phage display, reverse immunization, and next-generation sequencing-assisted antibody phage display to establish a highly myeloma-specific epitope fingerprint targeted by B-cell responses of 18 patients in clinical remission. We found that allogeneic HSCT more efficiently allowed production of myeloma-specific antibodies compared with autologous HSCT and that a highly reactive epitope recognition signature correlated with superior response to treatment. Next, we performed myeloma cell surface screenings of phage-displayed patient transplant immunomes. Although some of the screenings yielded clear-cut surface binders, the majority of screenings did not, suggesting that many of the targeted antigens may in fact not be accessible to the B-cell immune system in untreated myeloma cells. This fit well with the identification of heat-shock proteins as a class of antigens that showed overall the broadest reactivity with myeloma patient sera after allogeneic HSCT and that may be significantly translocated to the cell surface upon treatment as a result of immunogenic cell death. Our data reveal a disease-specific epitope signature of MM that is predictive for response to treatment. Mining of transplant immunomes for strong myeloma surface binders may open up avenues for myeloma immunotherapy., (© 2016 by The American Society of Hematology.)
- Published
- 2016
- Full Text
- View/download PDF
7. Functional autoantibodies against SSX-2 and NY-ESO-1 in multiple myeloma patients after allogeneic stem cell transplantation.
- Author
-
Luetkens T, Kobold S, Cao Y, Ristic M, Schilling G, Tams S, Bartels BM, Templin J, Bartels K, Hildebrandt Y, Yousef S, Marx A, Haag F, Bokemeyer C, Kröger N, and Atanackovic D
- Subjects
- Adult, Aged, B-Lymphocytes immunology, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Complement Activation, Enzyme-Linked Immunosorbent Assay, Epitopes chemistry, Female, Gene Expression Regulation, Neoplastic, Humans, K562 Cells, Male, Middle Aged, Multiple Myeloma therapy, Real-Time Polymerase Chain Reaction, Transplantation, Homologous, Antigens, Neoplasm immunology, Autoantibodies immunology, Hematopoietic Stem Cell Transplantation, Membrane Proteins immunology, Multiple Myeloma immunology, Neoplasm Proteins immunology, Repressor Proteins immunology
- Abstract
Background: Multiple myeloma (MM) is the malignancy with the most frequent expression of the highly immunogenic cancer-testis antigens (CTA), and we have performed the first analysis of longitudinal expression, immunological properties, and fine specificity of CTA-specific antibody responses in MM., Methods: Frequency and characteristics of antibody responses against cancer-testis antigens MAGE-A3, NY-ESO-1, PRAME, and SSX-2 were analyzed using peripheral blood (N = 1094) and bone marrow (N = 200) plasma samples from 194 MM patients., Results: We found that antibody responses against CTA were surprisingly rare, only 2.6 and 3.1 % of patients evidenced NY-ESO-1- and SSX-2-specific antibodies, respectively. NY-ESO-1-specific responses were observed during disease progression, while anti-SSX-2 antibodies appeared after allogeneic stem cell transplantation and persisted during clinical remission. We found that NY-ESO-1- and SSX-2-specific antibodies were both capable of activating complement and increasing CTA uptake by antigen-presenting cells. SSX-2-specific antibodies were restricted to IgG3, NY-ESO-1 responses to IgG1 and IgG3. Remarkably, NY-ESO-1-positive sera recognized various non-contiguous regions, while SSX-2-specific responses were directed against a single 6mer epitope, SSX-2(85-90)., Conclusions: We conclude that primary autoantibodies against intracellular MM-specific tumor antigens SSX-2 and NY-ESO-1 are rare but functional. While their contribution to disease control still remains unclear, our data demonstrate their theoretic ability to affect cellular anti-tumor immunity by formation and uptake of mono- and polyvalent immune complexes.
- Published
- 2014
- Full Text
- View/download PDF
8. Antigen-specificity of oligoclonal abnormal protein bands in multiple myeloma after allogeneic stem cell transplantation.
- Author
-
Rahlff J, Trusch M, Haag F, Bacher U, Horst A, Schlüter H, and Binder M
- Subjects
- Antigens, Neoplasm blood, Chaperonin 60 blood, Humans, Immunoglobulins blood, Immunoglobulins immunology, Mitochondrial Proteins blood, Neoplasm Proteins blood, Neoplasm Proteins immunology, Phosphopyruvate Hydratase blood, Phosphopyruvate Hydratase immunology, Proliferating Cell Nuclear Antigen blood, Proliferating Cell Nuclear Antigen immunology, Up-Regulation, Vimentin blood, Vimentin immunology, alpha-Glucosidases blood, alpha-Glucosidases immunology, Antigens, Neoplasm immunology, Bone Marrow Cells immunology, Chaperonin 60 immunology, Mitochondrial Proteins immunology, Multiple Myeloma immunology, Stem Cell Transplantation
- Abstract
Myeloma patients may develop oligoclonal immunoglobulins, so-called abnormal protein bands (APB), after stem cell transplantation. APB do not correspond to the patient's paraprotein and confer a good prognosis. We set out to investigate whether such APB represent a humoral anti-myeloma immune response by screening immunoglobulins of 15 myeloma patients after allogeneic stem cell transplantation and a control group of healthy donors for reactivity with myeloma protein extracts. While the immunoglobulins of healthy donors did not react with myeloma protein extracts, patient-derived immunoglobulins showed variable levels of interaction, depending on the presence of APB on immunofixation. Most commonly, we detected interactions with heat-shock proteins, followed by neutral alpha-glucosidase, alpha-enolase and vimentin, as well as proliferating cell nuclear antigen and MAGEA4. More than 80% of targets were upregulated in myeloma. Heat-shock protein 60 (HSP60) was subsequently evaluated as an exemplary antigen. We found that HSP60 was aberrantly displayed on the surface of primary myeloma cells. Indeed, patient-derived APB-containing immunoglobulins recognized surface HSP60 suggesting that this antigen becomes accessible to the immune system after aberrant membrane exposition. We conclude that immunoglobulin fractions with APB recognize recurrent myeloma antigens and that this humoral response may contribute to the more favorable prognosis in patients with APB.
- Published
- 2012
- Full Text
- View/download PDF
9. Longitudinal analysis of tetanus- and influenza-specific IgG antibodies in myeloma patients.
- Author
-
Kobold S, Luetkens T, Bartels BM, Cao Y, Hildebrandt Y, Sezer O, Reinhard H, Templin J, Bartels K, Lajmi N, Haag F, Bokemeyer C, Kröger N, and Atanackovic D
- Subjects
- Aged, Antibodies, Bacterial immunology, Antibodies, Viral immunology, Antineoplastic Agents administration & dosage, Antineoplastic Agents therapeutic use, Case-Control Studies, Enzyme-Linked Immunosorbent Assay, Female, Humans, Immunity, Humoral, Immunoglobulin G immunology, Immunoglobulins, Intravenous therapeutic use, Immunosuppression Therapy, Injections, Intravenous, Longitudinal Studies, Male, Middle Aged, Multiple Myeloma therapy, Nucleocapsid Proteins, RNA-Binding Proteins immunology, Stem Cell Transplantation, Transplantation, Homologous, Viral Core Proteins immunology, Antibodies, Bacterial biosynthesis, Antibodies, Viral biosynthesis, B-Lymphocytes immunology, Immunoglobulin G blood, Alphainfluenzavirus immunology, Multiple Myeloma immunology, Tetanus Toxoid immunology
- Abstract
Background: Multiple myeloma (MM) and its therapies may induce a severely compromised humoral immunity. We have performed a longitudinal analysis of IgG-antibody responses against influenza virus (FLU) and tetanus toxoid (TT) as surrogate markers for the B cell-mediated immunity in MM patients., Methods: 1094 serum samples of 190 MM patients and samples from 100 healthy donors were analyzed by ELISA for FLU- and TT-specific antibodies., Results: MM patients evidenced lower levels of FLU- and TT-specific antibodies than healthy controls (P < 0.001). Immunoreactivity decreased with progressing disease and worsening clinical status. Levels of FLU- and TT-specific antibodies increased shortly (0-6 months) after alloSCT (P < 0.001), a time-period during which intravenous immunoglobulin (IVIG) is routinely applied. Thereafter, antibody concentrations declined and remained suppressed for 3 years in the case of FLU-specific and for more than 5 years in the case of TT-specific antibodies., Conclusions: We found that MM is associated with a profound disease- and therapy-related immunosuppression, which is compensated for a few months after alloSCT, most likely by application of IVIG. This and the differences regarding the recovery of anti-FLU and anti-TT antibody titers during the following years need to be taken into account for optimizing IVIG application and immunization after alloSCT.
- Published
- 2012
- Full Text
- View/download PDF
10. Patients with multiple myeloma develop SOX2-specific autoantibodies after allogeneic stem cell transplantation.
- Author
-
Kobold S, Tams S, Luetkens T, Cao Y, Sezer O, Bartels BM, Reinhard H, Templin J, Bartels K, Hildebrandt Y, Lajmi N, Marx A, Haag F, Bokemeyer C, Kröger N, and Atanackovic D
- Subjects
- Antibody Specificity immunology, Autoantibodies blood, Cell Line, Tumor, Epitopes chemistry, Epitopes immunology, Humans, Immunoglobulin G blood, Immunoglobulin G immunology, Multiple Myeloma genetics, Prognosis, SOXB1 Transcription Factors genetics, SOXB1 Transcription Factors metabolism, Transplantation, Homologous, Autoantibodies immunology, Hematopoietic Stem Cell Transplantation, Multiple Myeloma immunology, Multiple Myeloma therapy, SOXB1 Transcription Factors immunology
- Abstract
The occurrence of SOX2-specific autoantibodies seems to be associated with an improved prognosis in patients with monoclonal gammopathy of undetermined significance (MGUS). However, it is unclear if SOX2-specific antibodies also develop in established multiple myeloma (MM). Screening 1094 peripheral blood (PB) sera from 196 MM patients and 100 PB sera from healthy donors, we detected SOX2-specific autoantibodies in 7.7% and 2.0% of patients and donors, respectively. We identified SOX2(211-230) as an immunodominant antibody-epitope within the full protein sequence. SOX2 antigen was expressed in most healthy tissues and its expression did not correlate with the number of BM-resident plasma cells. Accordingly, anti-SOX2 immunity was not related to SOX2 expression levels or tumor burden in the patients' BM. The only clinical factor predicting the development of anti-SOX2 immunity was application of allogeneic stem cell transplantation (alloSCT). Anti-SOX2 antibodies occurred more frequently in patients who had received alloSCT (n = 74). Moreover, most SOX2-seropositive patients had only developed antibodies after alloSCT. This finding indicates that alloSCT is able to break tolerance towards this commonly expressed antigen. The questions whether SOX2-specific autoantibodies merely represent an epiphenomenon, are related to graft-versus-host effects or participate in the immune control of myeloma needs to be answered in prospective studies.
- Published
- 2011
- Full Text
- View/download PDF
11. Mouse CD38-Specific Heavy Chain Antibodies Inhibit CD38 GDPR-Cyclase Activity and Mediate Cytotoxicity Against Tumor Cells.
- Author
-
Baum, Natalie, Eggers, Marie, Koenigsdorf, Julia, Menzel, Stephan, Hambach, Julia, Staehler, Tobias, Fliegert, Ralf, Kulow, Frederike, Adam, Gerhard, Haag, Friedrich, Bannas, Peter, and Koch-Nolte, Friedrich
- Subjects
MEMBRANE proteins ,IMMUNOGLOBULINS ,LABORATORY mice ,ANTIBODY-dependent cell cytotoxicity ,MULTIPLE myeloma - Abstract
CD38 is the major NAD
+ -hydrolyzing ecto-enzyme in most mammals. As a type II transmembrane protein, CD38 is also a promising target for the immunotherapy of multiple myeloma (MM). Nanobodies are single immunoglobulin variable domains from heavy chain antibodies that naturally occur in camelids. Using phage display technology, we isolated 13 mouse CD38-specific nanobodies from immunized llamas and produced these as recombinant chimeric mouse IgG2a heavy chain antibodies (hcAbs). Sequence analysis assigned these hcAbs to five distinct families that bind to three non-overlapping epitopes of CD38. Members of families 4 and 5 inhibit the GDPR-cyclase activity of CD38. Members of families 2, 4 and 5 effectively induce complement-dependent cytotoxicity against CD38-expressing tumor cell lines, while all families effectively induce antibody dependent cellular cytotoxicity. Our hcAbs present unique tools to assess cytotoxicity mechanisms of CD38-specific hcAbs in vivo against tumor cells and potential off-target effects on normal cells expressing CD38 in syngeneic mouse tumor models, i.e. in a fully immunocompetent background. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
12. Daratumumab and Nanobody-Based Heavy Chain Antibodies Inhibit the ADPR Cyclase but not the NAD + Hydrolase Activity of CD38-Expressing Multiple Myeloma Cells.
- Author
-
Baum, Natalie, Fliegert, Ralf, Bauche, Andreas, Hambach, Julia, Menzel, Stephan, Haag, Friedrich, Bannas, Peter, and Koch-Nolte, Friedrich
- Subjects
IMMUNOGLOBULINS ,HIGH performance liquid chromatography ,MONOCLONAL antibodies ,HYDROLASES ,GLYCOSIDASES ,GLYCOPROTEINS ,MULTIPLE myeloma ,CELL lines ,DRUG toxicity - Abstract
Simple Summary: Multiple myeloma is a hematological malignancy of antibody-producing plasma cells in the bone marrow. Nucleotides released from cells in the tumor microenvironment act as inflammatory danger signals. CD38 and other enzymes on the surface of cancer cells hydrolyze these nucleotides to immunosuppressive mediators, thereby hampering anti-tumor immune responses. Daratumumab and other CD38-specific antibodies mediate killing of tumor cells by natural killer cells, macrophages, and the complement system. Here, we investigated whether CD38-specific antibodies also inhibit the enzyme activity of CD38-expressing tumor cells, thereby providing a potential second mode of action. Our results showed that daratumumab and nanobody-based heavy chain antibodies inhibit the ADPR cyclase but not the NAD
+ hydrolase activity of CD38. Thus, there remains a need for better CD38-inhibitory antibodies. The nucleotides ATP and NAD+ are released from stressed cells as endogenous danger signals. Ecto-enzymes in the tumor microenvironment hydrolyze these inflammatory nucleotides to immunosuppressive adenosine, thereby, hampering anti-tumor immune responses. The NAD+ hydrolase CD38 is expressed at high levels on the cell surface of multiple myeloma (MM) cells. Daratumumab, a CD38-specific monoclonal antibody promotes cytotoxicity against MM cells. With long CDR3 loops, nanobodies and nanobody-based heavy chain antibodies (hcAbs) might bind to cavities on CD38 and thereby inhibit its enzyme activity more potently than conventional antibodies. The goal of our study was to establish assays for monitoring the enzymatic activities of CD38 on the cell surface of tumor cells and to assess the effects of CD38-specific antibodies on these activities. We monitored the enzymatic activity of CD38-expressing MM and other tumor cell lines, using fluorometric and HPLC assays. Our results showed that daratumumab and hcAb MU1067 inhibit the ADPR cyclase but not the NAD+ hydrolase activity of CD38-expressing MM cells. We conclude that neither clinically approved daratumumab nor recently developed nanobody-derived hcAbs provide a second mode of action against MM cells. Thus, there remains a quest for "double action" CD38-inhibitory antibodies. [ABSTRACT FROM AUTHOR]- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.