Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
BMC Cancer ; 21(1): 994, 2021 Sep 06.
Article in English | MEDLINE | ID: mdl-34488679

ABSTRACT

BACKGROUND: Many clinical trials have assessed the effect and safety of monoclonal antibodies (MAbs) in combination with proteasome inhibitors or immunomodulators plus dexamethasone/prednisone for the treatment of multiple myeloma (MM). The treatment outcomes of comparing different MAbs in combination with the above-mentioned agents remained unclear. We performed the meta-analysis to indirectly compare the effect and safety of MAbs targeting CD38, SLAMF7, and PD-1/PD-L1 in combination with bortezomib/immunomodulators plus dexamethasone/prednisone for patients with MM. METHODS: We searched thoroughly in the databases for randomised controlled trials (RCTs) in which at least one of the three MAbs were included. We included eleven eligible RCTs with 5367 patients in the meta-analysis. Statistical analysis was carried out using StataMP14 and Indirect Treatment Comparisons software. RESULTS: We calculated hazard ratios (HRs) for overall survival (OS) and progression-free survival (PFS) and relative risk (RR) for overall response rate, complete response (CR) or better, very good partial response (VGPR) or better, VGPR, partial response, stable disease, and grade 3 or higher adverse events among the three groups. The HRs for PFS of the CD38 group vs SLAMF7 group, CD38 group vs PD-1/PD-L1 group, and SLAMF7 group vs PD-1/PD-L1 group were 0.662 (95%CI 0.543-0.806), 0.317 (95%CI 0.221-0.454), and 0.479 (95%CI 0.328-0.699), respectively. The HR for OS of the CD38 group vs SLAMF7 group was 0.812 (95%CI 0.584-1.127). The RR for CR or better in the CD38 group vs SLAMF7 group was 2.253 (95%CI 1.284-3.955). The RR for neutropenia of the CD38 group vs SLAMF7 group was 1.818 (95%CI 1.41-2.344). CONCLUSIONS: Treatment with the CD38 group had longer PFS and better treatment response than that with the SLAMF7 or PD-1/PD-L1 group. In addition, the SLAMF7 group prolonged PFS compared with the PD-1/PD-L1 group and was associated with a lower incidence of grade 3 or higher neutropenia than the CD38 and PD-1/PD-L1 group. In conclusion, MAbs targeting CD38 are the best, followed by those targeting SLAMF7; MAbs targeting PD-1/PD-L1 are the worst when in combination with bortezomib/immunomodulators plus dexamethasone/prednisone for the treatment of MM.


Subject(s)
ADP-ribosyl Cyclase 1/antagonists & inhibitors , Antibodies, Monoclonal/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Membrane Glycoproteins/antagonists & inhibitors , Multiple Myeloma/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Signaling Lymphocytic Activation Molecule Family/antagonists & inhibitors , ADP-ribosyl Cyclase 1/immunology , B7-H1 Antigen/immunology , Bortezomib/administration & dosage , Dexamethasone/administration & dosage , Humans , Immunologic Factors/therapeutic use , Membrane Glycoproteins/immunology , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Prednisone/administration & dosage , Prognosis , Programmed Cell Death 1 Receptor/immunology , Randomized Controlled Trials as Topic , Signaling Lymphocytic Activation Molecule Family/immunology
2.
Cancer Sci ; 112(10): 3995-4004, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34310776

ABSTRACT

Multiple myeloma (MM) is a refractory plasma cell tumor. In myeloma cells, the transcription factor IRF4, the master regulator of plasma cells, is aberrantly upregulated and plays an essential role in oncogenesis. IRF4 forms a positive feedback loop with MYC, leading to additional tumorigenic properties. In recent years, molecular targeted therapies have contributed to a significant improvement in the prognosis of MM. Nevertheless, almost all patients experience disease progression, which is thought to be a result of treatment resistance induced by various elements of the bone marrow microenvironment. Among these, the hypoxic response, one of the key processes for cellular homeostasis, induces hypoxia-adapted traits such as undifferentiation, altered metabolism, and dissemination, leading to drug resistance. These inductions are caused by ectopic gene expression changes mediated by the activation of hypoxia-inducible factors (HIFs). By contrast, the expression levels of IRF4 and MYC are markedly reduced by hypoxic stress. Notably, an anti-apoptotic capability is usually acquired under both normoxic and hypoxic conditions, but the mechanism is distinct. This fact strongly suggests that myeloma cells may survive by switching their dependent regulatory factors from IRF4 and MYC (normoxic bone marrow region) to HIF (hypoxic bone marrow microenvironment). Therefore, to achieve deep remission, combination therapeutic agents, which are complementarily effective against both IRF4-MYC-dominant and HIF-dominated fractions, may become an important therapeutic strategy for MM.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interferon Regulatory Factors/metabolism , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Tumor Hypoxia/physiology , ADP-ribosyl Cyclase 1/antagonists & inhibitors , ADP-ribosyl Cyclase 1/metabolism , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Marrow Cells/physiology , Cell Dedifferentiation , Cell Hypoxia/physiology , Cell Movement/physiology , Cellular Microenvironment/physiology , Circulating MicroRNA/metabolism , Disease Progression , Drug Resistance, Neoplasm/physiology , Feedback, Physiological , Glycolysis/physiology , Hexokinase/metabolism , Homeostasis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunologic Factors/therapeutic use , Interferon Regulatory Factors/genetics , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/metabolism , Molecular Targeted Therapy/methods , Multiple Myeloma/etiology , Multiple Myeloma/genetics , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/physiology , Oxygen , Partial Pressure , Proteasome Inhibitors/therapeutic use , Proto-Oncogene Proteins c-myc/genetics , Signaling Lymphocytic Activation Molecule Family/antagonists & inhibitors , Up-Regulation
3.
Nat Commun ; 12(1): 1893, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33767202

ABSTRACT

B cells have essential functions in multiple sclerosis and in its mouse model, experimental autoimmune encephalomyelitis, both as drivers and suppressors of the disease. The suppressive effects are driven by a regulatory B cell (Breg) population that functions, primarily but not exclusively, via the production of IL-10. However, the mechanisms modulating IL-10-producing Breg abundance are poorly understood. Here we identify SLAMF5 for controlling IL-10+ Breg maintenance and function. In EAE, the deficiency of SLAMF5 in B cells causes accumulation of IL10+ Bregs in the central nervous system and periphery. Blocking SLAMF5 in vitro induces both human and mouse IL-10-producing Breg cells and increases their survival with a concomitant increase of a transcription factor, c-Maf. Finally, in vivo SLAMF5 blocking in EAE elevates IL-10+ Breg levels and ameliorates disease severity. Our results suggest that SLAMF5 is a negative moderator of IL-10+ Breg cells, and may serve as a therapeutic target in MS and other autoimmune diseases.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Interleukin-10/immunology , Signaling Lymphocytic Activation Molecule Family/metabolism , Animals , Cell Survival/immunology , Cells, Cultured , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Multiple Sclerosis/immunology , Signaling Lymphocytic Activation Molecule Family/antagonists & inhibitors , Signaling Lymphocytic Activation Molecule Family/genetics
4.
Clin Adv Hematol Oncol ; 19(3): 166-174, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33739965

ABSTRACT

Antibody therapy, which has become a critical option in the treatment of multiple myeloma (MM), includes monoclonal antibodies, antibody-drug conjugates, and bispecific antibodies. Anti-CD38 and anti-SLAMF7 monoclonal antibodies were the first to enter the MM portfolio as treatment options for relapsed/ refractory MM. More recently, daratumumab has become important in the treatment of newly diagnosed MM, and a subcutaneous formulation has been approved. BCMA-targeted antibody-drug conjugates and bispecific antibodies, which are the newest antibody therapies to be investigated, provide additional therapeutic options for patients with heavily pretreated MM. This article reviews how antibody therapy has influenced the treatment of MM, describes the unique adverse event profiles of each relevant drug class, and explains how to incorporate antibody therapy into practice.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Immunoconjugates/therapeutic use , Multiple Myeloma/drug therapy , ADP-ribosyl Cyclase 1/antagonists & inhibitors , ADP-ribosyl Cyclase 1/immunology , Animals , Antibodies, Bispecific/adverse effects , Antibodies, Bispecific/immunology , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/immunology , B-Cell Maturation Antigen/antagonists & inhibitors , B-Cell Maturation Antigen/immunology , Humans , Immunoconjugates/adverse effects , Immunoconjugates/immunology , Multiple Myeloma/immunology , Signaling Lymphocytic Activation Molecule Family/antagonists & inhibitors , Signaling Lymphocytic Activation Molecule Family/immunology
5.
Mol Ther ; 29(2): 702-717, 2021 02 03.
Article in English | MEDLINE | ID: mdl-33129371

ABSTRACT

Chimeric antigen receptors (CARs) are fusion proteins that contain antigen-recognition domains and T cell signaling domains. Signaling lymphocytic-activation molecule F7 (SLAMF7) is a promising target for CAR T cell therapies of the plasma cell malignancy multiple myeloma (MM) because SLAMF7 is expressed by MM but not normal nonhematopoietic cells. We designed CARs targeting SLAMF7. We transduced human T cells with anti-SLAMF7 CARs containing either CD28 or 4-1BB costimulatory domains. T cells expressing CD28-containing CARs or 4-1BB-containing CARs recognized SLAMF7 in vitro. SLAMF7-specific cytokine release was higher for T cells expressing CARs with CD28 versus 4-1BB domains. In murine solid tumor and disseminated tumor models, anti-tumor activity of T cells was superior with CD28-containing CARs versus 4-1BB-containing CARs. Because of SLAMF7 expression on some normal leukocytes, especially natural killer cells that control certain viral infections, the inclusion of a suicide gene with an anti-SLAMF7 CAR is prudent. We designed a construct with a CD28-containing anti-SLAMF7 CAR and a suicide gene. The suicide gene encoded a dimerization domain fused to a caspase-9 domain. T cells expressing the anti-SLAMF7 CAR plus suicide-gene construct specifically recognized SLAMF7 in vitro and eliminated tumors from mice. T cells expressing this construct were eliminated on demand by administering the dimerizing agent AP1903 (rimiducid).


Subject(s)
Gene Expression , Genes, Transgenic, Suicide/genetics , Immunotherapy, Adoptive , Receptors, Chimeric Antigen/immunology , Signaling Lymphocytic Activation Molecule Family/antagonists & inhibitors , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Animals , Disease Models, Animal , Humans , Immunotherapy, Adoptive/methods , Mice , Multiple Myeloma/immunology , Multiple Myeloma/therapy , Receptors, Chimeric Antigen/genetics , Signaling Lymphocytic Activation Molecule Family/immunology , Xenograft Model Antitumor Assays
6.
J Immunother Cancer ; 8(1)2020 03.
Article in English | MEDLINE | ID: mdl-32217758

ABSTRACT

BACKGROUND: Developing novel strategies to overcome the immunosuppressive tumor microenvironment is a critically important area of cancer therapy research. Here, we assess the therapeutic potential of CD244 (2B4/signaling lymphocyte activation molecule family 4), an immunoregulatory receptor found on a variety of immune cells, including exhausted CD8+ T cells, dendritic cells (DCs), and myeloid-derived suppressor cells (MDSCs). METHODS: Using de-identified human tumor and blood samples from patients with head and neck squamous cell carcinoma (HNSCC) and HNSCC models in WT and CD244-/- mice, we assessed the therapeutic potential of CD244 using flow cytometry, RT-PCR, Luminex immunoassays and histopathological analyses. RESULTS: Compared with healthy tissues, tumor infiltrating CD8+ T cells from HNSCC patients and a HNSCC mouse model showed significant increased expression of CD244 expression that correlated with PD1 expression. Moreover, CD244 was increased on intratumoral DC and MDSC and high CD244 expression correlated with PD-L1 expression and increased spontaneous expression of immune-suppressive mediators. In addition, CD244 activation inhibited production of proinflammatory cytokines in human DC in vitro. Importantly, CD244-/- mice showed significantly impaired tumor growth of HNSCC and interventional treatment of WT mice with anti-CD244 monoclonal antibody significantly impaired the growth of established HNSCC tumors and increased tumor-infiltrating CD8+ T cells. CONCLUSIONS: Together these data suggest that CD244 contributes to the overall immune-suppressive environment and therefore has potential as a new immunotherapy target in the treatment of malignancies.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD8-Positive T-Lymphocytes/immunology , Head and Neck Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Signaling Lymphocytic Activation Molecule Family/antagonists & inhibitors , Signaling Lymphocytic Activation Molecule Family/metabolism , Squamous Cell Carcinoma of Head and Neck/immunology , Animals , Case-Control Studies , Cell Line, Tumor , Dendritic Cells/immunology , Disease Models, Animal , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Humans , Leukocytes, Mononuclear/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Targeted Therapy , Myeloid-Derived Suppressor Cells/immunology , Signaling Lymphocytic Activation Molecule Family/genetics , Signaling Lymphocytic Activation Molecule Family/immunology , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology , Tumor Microenvironment/immunology
7.
Clin Cancer Res ; 26(10): 2308-2317, 2020 05 15.
Article in English | MEDLINE | ID: mdl-31969330

ABSTRACT

PURPOSE: ABBV-838 is an antibody-drug conjugate targeting a unique epitope of CD2 subset 1, a cell-surface glycoprotein expressed on multiple myeloma cells. This phase I/Ib first-in-human, dose-escalation study (trial registration ID: NCT02462525) evaluated the safety, pharmacokinetics, and preliminary activity of ABBV-838 in patients with relapsed and refractory multiple myeloma (RRMM). PATIENTS AND METHODS: Eligible patients (≥18 years) received ABBV-838 (3+3 design) intravenously starting from 0.6 mg/kg up to 6.0 mg/kg for 3-week dosing intervals (Q3W). Patients could continue ABBV-838 for up to 24 months. Assessment of alternate dosing intervals (Q1W and Q2W) was conducted in parallel. RESULTS: As of March 2017, 75 patients received at least one dose of ABBV-838. The most common any-grade treatment-emergent adverse events (TEAE) were neutropenia and anemia (28.0% each), fatigue (26.7%), and nausea (25.3%). Grade 3/4/5 TEAEs were reported in 73.3% of patients across all treatment groups; most common were neutropenia (20.0%), anemia (18.7%), and leukopenia (13.3%). Grade 3/4/5 ABBV-838-related TEAEs were reported by 40.0% of patients across all treatment groups. Overall, 4.0% of patients experienced TEAEs leading to death, none ABBV-838 related. The MTD was not reached; the selected recommended dose for the expansion cohort was 5.0 mg/kg Q3W. Pharmacokinetic analysis showed that exposure was approximately dose proportional. The overall response rate was 10.7%; very good partial responses and partial responses were achieved by 2 (2.7%) and 6 (8.0%) patients, respectively. CONCLUSIONS: These results demonstrate that ABBV-838 is safe and well-tolerated in patients with RRMM with a very limited efficacy.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Immunoconjugates/therapeutic use , Multiple Myeloma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Signaling Lymphocytic Activation Molecule Family/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/pharmacokinetics , Cohort Studies , Drug Resistance, Neoplasm , Female , Follow-Up Studies , Humans , Immunoconjugates/pharmacokinetics , Male , Middle Aged , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Prognosis , Salvage Therapy , Signaling Lymphocytic Activation Molecule Family/immunology , Tissue Distribution
8.
Clin Adv Hematol Oncol ; 18(11): 736-748, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33406065

ABSTRACT

Despite continued and considerable progress following the introduction of proteasome inhibitors and immunomodulatory agents, multiple myeloma (MM) remains an incurable disease, and new therapeutic strategies are urgently needed. Monoclonal antibodies represent a well-established targeted approach to the treatment of MM, with selective killing properties and limited off-target toxicity. Since their approval, the anti-CD38 agent daratumumab, the anti-SLAMF7 agent elotuzumab, and most recently the anti-CD38 agent isatuximab have led to pivotal improvements in the treatment of double-refractory MM; currently, they are on their way to becoming integral parts in the up-front care of patients who have newly diagnosed MM, with daratumumab already approved in this setting. Several other antibody-based strategies are undergoing clinical assessment in MM. Although the investigation of checkpoint inhibitors in MM has been halted, bispecific T-cell engagers and especially antibody-drug conjugates demonstrate encouraging efficacy and manageable toxicity in triple class-refractory MM. The accelerated approval of belantamab mafodotin represents an important milestone in antibody development; its ability to target B-cell maturation antigen (BCMA) in advanced disease is now established. Here, we present an overview of the currently available monoclonal antibody treatments in MM and discuss the clinical value, significant potential, and possible limitations of these immunotherapeutic approaches to driving deeper responses and achieving longer overall survival among patients with a challenging disease.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Multiple Myeloma/drug therapy , ADP-ribosyl Cyclase 1/antagonists & inhibitors , ADP-ribosyl Cyclase 1/immunology , Antineoplastic Agents, Immunological/immunology , B-Cell Maturation Antigen/antagonists & inhibitors , B-Cell Maturation Antigen/immunology , Humans , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/immunology , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/immunology , Signaling Lymphocytic Activation Molecule Family/antagonists & inhibitors , Signaling Lymphocytic Activation Molecule Family/immunology
9.
JCI Insight ; 4(22)2019 11 14.
Article in English | MEDLINE | ID: mdl-31723059

ABSTRACT

In addition to its well-known beneficial effects for the treatment of several types of cancer, PD-1 blockade has shown encouraging results in preclinical models of sepsis and in a recent clinical trial in sepsis. Because cancer is the most common comorbidity in septic patients, here we aimed to determine the efficacy of PD-1 checkpoint blockade in the setting of sepsis complicated with preexisting malignancy. In a model of established lung cancer followed by cecal ligation and puncture-induced (CLP-induced) sepsis, PD-1 blockade exhibited no therapeutic effect on sepsis survival. This diminished efficacy of PD-1 blockade in cancer septic animals (septic animals with cancer) was characterized by a reduction in both the quality and quantity of PD-1+ responder cells. Specifically, CD8+ T cells isolated from cancer septic animals exhibited decreased CD28 expression and a reduction in the CXCR5+PD-1+ subset. In addition, flow cytometric analysis of T cells isolated from cancer septic animals revealed 2B4 as another possible checkpoint under these conditions. Administration of anti-2B4 to cancer septic animals significantly improved sepsis survival and was associated with increased T cell costimulatory receptor expression and decreased coinhibitory receptor expression. These results illustrate functions of coinhibitory receptors in the setting of sepsis complicated with cancer.


Subject(s)
Lung Neoplasms , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Sepsis , Signaling Lymphocytic Activation Molecule Family/antagonists & inhibitors , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/metabolism , Signaling Lymphocytic Activation Molecule Family/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
11.
Cancer Immunol Res ; 7(10): 1633-1646, 2019 10.
Article in English | MEDLINE | ID: mdl-31431433

ABSTRACT

Elotuzumab (Elo) is an IgG1 monoclonal antibody targeting SLAMF7 (CS1, CRACC, and CD319), which is highly expressed on multiple myeloma (MM) cells, natural killer (NK) cells, and subsets of other leukocytes. By engaging with FcγRIIIA (CD16), Elo promotes potent NK cell-mediated antibody-dependent cellular cytotoxicity (ADCC) and macrophage-mediated antibody-dependent cellular phagocytosis (ADCP) toward SLAMF7+ MM tumor cells. Relapsed/refractory MM patients treated with the combination of Elo, lenalidomide, and dexamethasone have improved progression-free survival. We previously showed that Elo enhances NK cell activity via a costimulation mechanism, independent of CD16 binding. Here, we further studied the effect of Elo on cytotoxicity of CD16-negative NK-92 cells. Elo, but not other SLAMF7 antibodies, uniquely enhanced cytotoxicity mediated by CD16-negative NK-92 cells toward SLAMF7+ target cells. Furthermore, this CD16-independent enhancement of cytotoxicity required expression of SLAMF7 containing the full cytoplasmic domain in the NK cells, implicating costimulatory signaling. The CD16-independent costimulation by Elo was associated with increased expression of NKG2D, ICAM-1, and activated LFA-1 on NK cells, and enhanced cytotoxicity was partially reduced by NKG2D blocking antibodies. In addition, an Fc mutant form of Elo that cannot bind CD16 promoted cytotoxicity of SLAMF7+ target cells by NK cells from most healthy donors, especially if previously cultured in IL2. We conclude that in addition to promoting NK cell-mediated ADCC (CD16-dependent) responses, Elo promoted SLAMF7-SLAMF7 interactions in a CD16-independent manner to enhance NK cytotoxicity toward MM cells.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Antibody-Dependent Cell Cytotoxicity , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Multiple Myeloma/drug therapy , Signaling Lymphocytic Activation Molecule Family/metabolism , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Killer Cells, Natural/drug effects , Lymphocyte Activation/drug effects , Multiple Myeloma/immunology , Multiple Myeloma/metabolism , Signaling Lymphocytic Activation Molecule Family/antagonists & inhibitors , Signaling Lymphocytic Activation Molecule Family/immunology
13.
Expert Rev Hematol ; 12(7): 481-496, 2019 07.
Article in English | MEDLINE | ID: mdl-31125526

ABSTRACT

Introduction: Multiple myeloma (MM) is characterized by the high tendency to relapse and develop drug resistance. Areas covered: This review focused on the main novel targets identified to design drugs for the treatment of relapsing MM patients. CD38 and SLAMF7 are the main surface molecules leading to the development of monoclonal antibodies (mAbs) recently approved for the treatment of relapsing MM patients. B cell maturation antigen (BCMA) is a suitable target for antibody-drug conjugates, bispecific T cell engager mAbs and Chimeric Antigen Receptor (CAR)-T cells. Moreover, the programmed cell death protein 1 (PD)-1/PD-Ligand (PD-L1) expression profile by MM cells and their microenvironment and the use of immune checkpoints inhibitors in MM patients are reported. Finally, the role of histone deacetylase (HDAC), B cell lymphoma (BCL)-2 family proteins and the nuclear transport protein exportin 1 (XPO1) as novel targets are also underlined. The clinical results of the new inhibitors in relapsing MM patients are discussed. Expert opinion: CD38, SLAMF7, and BCMA are the main targets for different immunotherapeutic approaches. Selective inhibitors of HDAC6, BCL-2, and XPO1 are new promising compounds under clinical investigation in relapsing MM patients.


Subject(s)
Biomarkers, Tumor , Molecular Targeted Therapy , Multiple Myeloma/etiology , Multiple Myeloma/therapy , ADP-ribosyl Cyclase 1/antagonists & inhibitors , ADP-ribosyl Cyclase 1/metabolism , B-Cell Maturation Antigen/antagonists & inhibitors , B-Cell Maturation Antigen/metabolism , Clinical Trials as Topic , Combined Modality Therapy , Humans , Immunomodulation/drug effects , Immunotherapy, Adoptive , Molecular Targeted Therapy/adverse effects , Molecular Targeted Therapy/methods , Multiple Myeloma/mortality , Multiple Myeloma/pathology , Recurrence , Signal Transduction/drug effects , Signaling Lymphocytic Activation Molecule Family/antagonists & inhibitors , Signaling Lymphocytic Activation Molecule Family/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Treatment Outcome , Tumor Microenvironment/drug effects
14.
Cancer Immunol Res ; 6(7): 776-787, 2018 07.
Article in English | MEDLINE | ID: mdl-29769244

ABSTRACT

Multiple myeloma (MM) is an incurable hematologic malignancy of plasma cells, with an estimated 30,000 new cases diagnosed each year in the United States, signifying the need for new therapeutic approaches. We hypothesized that targeting MM using a bispecific antibody (biAb) to simultaneously engage both innate and adaptive cytolytic immune cells could present potent antitumor activity. We engineered a biAb by fusing an anti-CS1 single-chain variable fragment (scFv) and an anti-NKG2D scFv (CS1-NKG2D biAb). Although NKG2D is a potent activation receptor ubiquitously expressed on mostly cytolytic immune cells including NK cells, CD8+ T cells, γδ T cells, and NKT cells, the CS1 tumor-associated antigen on MM represents a promising target. CS1-NKG2D biAb engaged human MM cell lines and NKG2D+ immune cells, forming immune synapses. In effector cells, CS1-NKG2D biAb triggered the phosphorylation of AKT, a downstream protein kinase of the activated NKG2D-DAP10 complex. The EC50 values of CS1-NKG2D biAb for CS1high and for CS1low MM cell lines with effector PBMCs were 10-12 and 10-9 mol/L, respectively. CS1-NKG2D biAb acted through multiple types of immune cells, and this induced cytotoxicity was both CS1- and NKG2D-specific. In vivo, survival was significantly prolonged using CS1-NKG2D biAb in a xenograft NOD-SCIDIL2γc-/- (NSG) mouse model engrafted with both human PBMCs and MM cell lines. Collectively, we demonstrated that the CS1-NKG2D biAb facilitated an enhanced immune synapse between CS1+ MM cells and NKG2D+ cytolytic innate and antigen-specific effector cells, which, in turn, activated these immune cells for improved clearance of MM. Cancer Immunol Res; 6(7); 776-87. ©2018 AACR.


Subject(s)
Antibodies, Bispecific/pharmacology , Cytotoxicity, Immunologic , Multiple Myeloma/immunology , NK Cell Lectin-Like Receptor Subfamily K/antagonists & inhibitors , Signaling Lymphocytic Activation Molecule Family/antagonists & inhibitors , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , Animals , Biomarkers , Cell Line, Tumor , Female , Humans , Immunological Synapses , Immunophenotyping , Interferon-gamma/metabolism , Mice , Multiple Myeloma/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , T-Lymphocyte Subsets/metabolism
15.
Autoimmun Rev ; 17(7): 674-682, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29729453

ABSTRACT

The signaling lymphocytic activation molecule (SLAM) family is comprised of nine distinct receptors (SLAMF1 through SLAMF9) that are expressed on hematopoietic cells. All of these receptors, with the exception of SLAMF4, are homotypic by nature as downstream signaling occurs when hematopoietic cells that express the same SLAM receptor interact. The SLAM family receptor function is largely controlled via SLAM associated protein (SAP) family adaptors. The SAP family adaptors consist of SAP, Ewing sarcoma associated transcript (EAT)-2, and EAT-2-related transducer (ERT). These adaptors associate with the cytoplasmic domain of the SLAM family receptors through phosphorylated tyrosines. Defects in SLAM family members and SAP adaptors have been implicated in causing immune deficiencies. This is exemplified in patients with X-linked lymphoproliferative (XLP) disease, where SAP undergoes a loss of function mutation. Furthermore, evidence has been accumulating that SLAM family members are potential targets for inflammatory and autoimmune diseases. This review will discuss the structure and function of the SLAM family receptors and SAP family adaptors, their role in immune regulation, and potential approaches to target this family of receptors therapeutically.


Subject(s)
Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Signaling Lymphocytic Activation Molecule Family/antagonists & inhibitors , Signaling Lymphocytic Activation Molecule Family/immunology , Animals , Antigens, CD/immunology , Humans , Immunity , Signal Transduction
17.
Expert Rev Hematol ; 11(5): 423-435, 2018 05.
Article in English | MEDLINE | ID: mdl-29582696

ABSTRACT

INTRODUCTION: the high expression of a number of surface antigens on malignant plasma cells, the bone marrow micro-environment and immune effector T cells, makes these appealing targets for immune therapy with monoclonal antibodies (mAbs). Areas covered: Two mAbs, anti-CD38 daratumumab (Dara) and anti-SLAMF7 elotuzumab (Elo), have achieved recent regulatory approval for relapsed or refractory MM (RRMM) and are currently being explored as possible treatment options in novel combinations and different settings. This review discusses the current landscape and possible development of anti-CD38 and anti-SLAMF7 mAbs. Expert commentary: Three phase III trials demonstrated a significant advantage in terms of response and PFS when Dara or Elo are combined with lenalidomide-dexamethsone (Rd) or bortezomib-dexamethsone (Vd), in comparison to doublet regimens, for patients with RRMM. Treatment algorithms including Dara- or Elo-based triplets may be defined on the basis of disease and patients' characteristics, as well as of their prior exposure to different classes of novel agents. Evaluation of these agents in new combination regimens, including second and third generation PIs and IMiDs, are under investigation. Moreover, use of mAbs in phases of the disease where the immune system is less compromised, such as newly diagnosed MM or even high-risk smoldering myeloma, appears logical.


Subject(s)
ADP-ribosyl Cyclase 1/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Immunotherapy/methods , Membrane Glycoproteins/antagonists & inhibitors , Multiple Myeloma/therapy , Neoplasm Proteins/antagonists & inhibitors , Signaling Lymphocytic Activation Molecule Family/antagonists & inhibitors , ADP-ribosyl Cyclase 1/metabolism , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Bortezomib/therapeutic use , Dexamethasone/therapeutic use , Humans , Lenalidomide/therapeutic use , Membrane Glycoproteins/metabolism , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Neoplasm Proteins/metabolism , Signaling Lymphocytic Activation Molecule Family/metabolism
18.
Leuk Lymphoma ; 59(4): 983-987, 2018 04.
Article in English | MEDLINE | ID: mdl-28782413
20.
Am Soc Clin Oncol Educ Book ; 37: 561-568, 2017.
Article in English | MEDLINE | ID: mdl-28561703

ABSTRACT

Multiple myeloma (MM) is a plasma cell malignancy characterized by the growth of tumor cells in the bone marrow. Properties of the tumor microenvironment provide both potential tumor-promoting and tumor-restricting properties. Targeting underlying immune triggers for evolution of tumors as well as direct attack of malignant plasma cells is an emerging focus of therapy for MM. The monoclonal antibodies daratumumab and elotuzumab, which target the plasma cell surface proteins CD38 and SLAMF7/CS1, respectively, particularly when used in combination with immunomodulatory agents and proteasome inhibitors, have resulted in high response rates and improved survival for patients with relapsed and refractory MM. A number of other monoclonal antibodies are in various stages of clinical development, including those targeting MM cell surface antigens, the bone marrow microenvironment, and immune effector T cells such as antiprogrammed cell death protein 1 antibodies. Bispecific preparations seek to simultaneously target MM cells and activate endogenous T cells to enhance efficacy. Cellular immunotherapy seeks to overcome the limitations of the endogenous antimyeloma immune response through adoptive transfer of immune effector cells with MM specificity. Allogeneic donor lymphocyte infusion can be effective but can cause graft-versus-host disease. The most promising approach appears to be genetically modified cellular therapy, in which T cells are given novel antigen specificity through expression of transgenic T-cell receptors (TCRs) or chimeric antigen receptors (CARs). CAR T cells against several different targets are under investigation in MM. Infusion of CD19-targeted CAR T cells following salvage autologous stem cell transplantation (SCT) was safe and extended remission duration in a subset of patients with relapsed/refractory MM. CAR T cells targeting B-cell maturation antigen (BCMA) appear most promising, with dramatic remissions seen in patients with highly refractory disease in three ongoing trials. Responses are associated with degree of CAR T-cell expansion/persistence and often toxicity, including cytokine release syndrome (CRS) and neurotoxicity. Ongoing and future studies are exploring correlates of response, ways to mitigate toxicity, and "universal" CAR T cells.


Subject(s)
Hematologic Neoplasms/genetics , Hematologic Neoplasms/therapy , Immunotherapy , Stem Cell Transplantation , ADP-ribosyl Cyclase 1/antagonists & inhibitors , ADP-ribosyl Cyclase 1/genetics , Antibodies, Monoclonal/therapeutic use , Combined Modality Therapy , Hematologic Neoplasms/immunology , Hematologic Neoplasms/pathology , Humans , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Plasma Cells/drug effects , Plasma Cells/pathology , Receptors, Antigen, T-Cell/antagonists & inhibitors , Signaling Lymphocytic Activation Molecule Family/antagonists & inhibitors , Signaling Lymphocytic Activation Molecule Family/genetics , T-Lymphocytes/immunology , Tumor Microenvironment/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...