Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 151
Filtrer
1.
Expert Opin Biol Ther ; 24(5): 365-381, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38757726

RÉSUMÉ

INTRODUCTION: Gain/amplification of 1q (+1q) represents one of the most prevalent cytogenetic abnormalities (CAs) observed in multiple myeloma (MM). Historical studies predating the advent of anti-CD38 monoclonal antibodies (moAbs) implicated + 1q in poor prognoses, prompting its integration into novel staging systems. However, with the emergence of daratumumab and isatuximab, two pivotal anti-CD38 moAbs, the landscape of MM therapy has undergone a profound transformation. AREAS COVERED: This review encompasses a comprehensive analysis of diverse study methodologies, including observational investigations, clinical trials, meta-analyses, and real-world database analyses. By synthesizing these data sources, we aim to provide an overview of the current understanding of + 1q in the context of anti-CD38 moAbs therapies. EXPERT OPINION: Despite the paucity of available data, evidence suggests a potential mitigating effect of daratumumab on the adverse prognostic implications of + 1q. However, this benefit seems to diminish in patients harboring ≥ 4 copies or with concurrent high-risk CAs. On the other hand, isatuximab demonstrated promising outcomes in the relapsed-refractory setting for + 1q MM patients. Nevertheless, direct comparison between the two compounds is currently challenging. The current evidence firmly supports the integration of anti-CD38 moAb-based therapies as the standard of care for + 1q patients, pending further elucidation.


Sujet(s)
Antigènes CD38 , Anticorps monoclonaux , Myélome multiple , Humains , Myélome multiple/traitement médicamenteux , Myélome multiple/génétique , Myélome multiple/immunologie , Antigènes CD38/immunologie , Antigènes CD38/antagonistes et inhibiteurs , Antigènes CD38/génétique , Anticorps monoclonaux/usage thérapeutique , Chromosomes humains de la paire 1/génétique , Glycoprotéines membranaires , Anticorps monoclonaux humanisés
2.
Front Immunol ; 15: 1382977, 2024.
Article de Anglais | MEDLINE | ID: mdl-38799465

RÉSUMÉ

CD38 antigen is a glycoprotein that found on the surface of several immune cells, and this property makes its monoclonal antibodies have the effect of targeted elimination of immune cells. Therefore, the CD38 monoclonal antibody (such as daratumumab, Isatuximab) becomes a new treatment option for membranous nephropathy, lupus nephritis, renal transplantation, and other refractory kidney diseases. This review summarizes the application of CD38 monoclonal antibodies in different kidney diseases and highlights future prospects.


Sujet(s)
Antigènes CD38 , Anticorps monoclonaux , Maladies du rein , Humains , Antigènes CD38/immunologie , Antigènes CD38/antagonistes et inhibiteurs , Antigènes CD38/métabolisme , Anticorps monoclonaux/usage thérapeutique , Maladies du rein/immunologie , Animaux , Glycoprotéines membranaires/immunologie , Glycoprotéines membranaires/antagonistes et inhibiteurs , Transplantation rénale , Anticorps monoclonaux humanisés/usage thérapeutique
3.
Cells ; 13(10)2024 May 20.
Article de Anglais | MEDLINE | ID: mdl-38786100

RÉSUMÉ

Current treatment strategies for multiple myeloma (MM) are highly effective, but most patients develop relapsed/refractory disease (RRMM). The anti-CD38/CD3xCD28 trispecific antibody SAR442257 targets CD38 and CD28 on MM cells and co-stimulates CD3 and CD28 on T cells (TCs). We evaluated different key aspects such as MM cells and T cells avidity interaction, tumor killing, and biomarkers for drug potency in three distinct cohorts of RRMM patients. We found that a significantly higher proportion of RRMM patients (86%) exhibited aberrant co-expression of CD28 compared to newly diagnosed MM (NDMM) patients (19%). Furthermore, SAR442257 mediated significantly higher TC activation, resulting in enhanced MM killing compared to bispecific functional knockout controls for all relapse cohorts (Pearson's r = 0.7). Finally, patients refractory to anti-CD38 therapy had higher levels of TGF-ß (up to 20-fold) compared to other cohorts. This can limit the activity of SAR442257. Vactoserib, a TGF-ß inhibitor, was able to mitigate this effect and restore sensitivity to SAR442257 in these experiments. In conclusion, SAR442257 has high potential for enhancing TC cytotoxicity by co-targeting CD38 and CD28 on MM and CD3/CD28 on T cells.


Sujet(s)
Antigènes CD38 , Myélome multiple , Lymphocytes T , Humains , Myélome multiple/traitement médicamenteux , Myélome multiple/anatomopathologie , Myélome multiple/immunologie , Antigènes CD38/métabolisme , Antigènes CD38/antagonistes et inhibiteurs , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Lymphocytes T/effets des médicaments et des substances chimiques , Antigènes CD3/métabolisme , Antigène CD28/métabolisme , Anticorps bispécifiques/pharmacologie , Anticorps bispécifiques/usage thérapeutique , Lignée cellulaire tumorale , Récidive
4.
Front Immunol ; 15: 1410457, 2024.
Article de Anglais | MEDLINE | ID: mdl-38765013

RÉSUMÉ

Introduction: CM313 is currently under clinical investigation for treatments of multiple myeloma, systemic lupus erythematosus, and immune thrombocytopenia. We aimed to report the preclinical profile of the novel therapeutic anti-CD38 monoclonal antibody (mAb) CM313, with an emphasis on the difference with other CD38-targeting mAb. Methods: The binding of CM313 to CD38 recombinant protein across species was assessed using ELISA. The binding of CM313 to CD38-positive (CD38+) cells was detected using flow cytometry assays. CM313-induced complement-dependent cytotoxicity (CDC), antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP) and apoptosis on different CD38+ cells were assessed by LDH release assays or flow cytometry assays. The effect of CM313 on CD38 enzymatic activity was measured using fluorescence spectroscopy. CM313 immunotoxicity in human blood was assessed using flow cytometry assays, ELISA, and LDH release assays. Anti-tumor activity of CM313 was assessed in multiple mouse xenograft models. Safety profile of CM313 were evaluated in cynomolgus monkeys and human CD38 transgenic (B-hCD38) mice. Results: There exist unique sequences at complementarity-determining regions (CDR) of CM313, which facilitates its affinity to CD38 is consistently higher across a spectrum of CD38+ cell lines than daratumumab. In vitro studies showed that CM313 induces comparable killing activity than daratumumab, including ADCC, CDC, ADCP, apoptosis induced by Fc-mediated cross-linking, and effectively inhibited the enzymatic activity of CD38. However, CM313 showed more potent CDC than isatuximab. In vivo, CM313 dose-dependently inhibited xenograft tumor growth, both as a monotherapy and in combination with dexamethasone or lenalidomide. Furthermore, CM313 was well tolerated with no drug-related clinical signs or off-target risks, as evidenced by 4-week repeat-dose toxicology studies in cynomolgus monkeys and B-hCD38 mice, with the later study showing no observed adverse effect level (NOAEL) of 300mg/kg once weekly. Discussion: CM313 is a novel investigational humanized mAb with a distinct CDR sequence, showing comparable killing effects with daratumumab and stronger CDC activity than isatuximab, which supports its clinical development.


Sujet(s)
Antigènes CD38 , Anticorps monoclonaux , Cytotoxicité à médiation cellulaire dépendante des anticorps , Macaca fascicularis , Animaux , Antigènes CD38/immunologie , Antigènes CD38/antagonistes et inhibiteurs , Humains , Souris , Anticorps monoclonaux/pharmacologie , Anticorps monoclonaux/immunologie , Cytotoxicité à médiation cellulaire dépendante des anticorps/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Tests d'activité antitumorale sur modèle de xénogreffe , Femelle , Souris transgéniques , Apoptose/effets des médicaments et des substances chimiques , Antinéoplasiques immunologiques/pharmacologie , Glycoprotéines membranaires
5.
Front Immunol ; 15: 1346178, 2024.
Article de Anglais | MEDLINE | ID: mdl-38680487

RÉSUMÉ

Introduction: Natural killer/T cell lymphoma (NKTL) is an aggressive malignancy associated with poor prognosis. This is largely due to limited treatment options, especially for relapsed patients. Immunotherapies like immune checkpoint inhibitors (ICI) and anti-CD38 therapies have shown promising but variable clinical efficacies. Combining these therapies has been suggested to enhance efficacy. Methods: We conducted a case study on a relapsed NKTL patient treated sequentially with anti-CD38 followed by ICI (anti-PD1) using cytometry analyses. Results and Discussion: Our analysis showed an expected depletion of peripheral CD38+ B cells following anti-CD38 treatment. Further analysis indicated that circulating anti-CD38 retained their function for up to 13 weeks post-administration. Anti-PD1 treatment triggered re-activation and upregulation of CD38 on the T cells. Consequently, these anti-PD1-activated T cells were depleted by residual circulating anti-CD38, rendering the ICI treatment ineffective. Finally, a meta-analysis confirmed this counterproductive effect, showing a reduced efficacy in patients undergoing combination therapy. In conclusion, our findings demonstrate that sequential anti-CD38 followed by anti-PD1 therapy leads to a counterproductive outcome in NKTL patients. This suggests that the treatment sequence is antithetic and warrants re-evaluation for optimizing cancer immunotherapy strategies.


Sujet(s)
Antigènes CD38 , Inhibiteurs de points de contrôle immunitaires , Humains , Antigènes CD38/antagonistes et inhibiteurs , Antigènes CD38/métabolisme , Antigènes CD38/immunologie , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Lymphome T-NK extraganglionnaire/thérapie , Lymphome T-NK extraganglionnaire/immunologie , Lymphome T-NK extraganglionnaire/traitement médicamenteux , Glycoprotéines membranaires/antagonistes et inhibiteurs , Mâle , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Adulte d'âge moyen , Femelle , Résultat thérapeutique
6.
Clin Lymphoma Myeloma Leuk ; 24(6): 358-363, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38519329

RÉSUMÉ

Isatuximab-based combinations are among the accepted standard-of-care regimens for early-line treatment of patients with relapsed/refractory multiple myeloma (RRMM), based on the results of the Phase 3 ICARIA-MM and IKEMA trials. Further study findings have shown benefit with Isa-based combinations in patients with newly diagnosed MM, as reported from the randomized GMMG-HD7 and CONCEPT trials. Isa is currently approved in various countries for intravenous (IV) administration in patients with RRMM. A more convenient route of administration, such as subcutaneous (SC) injection, and faster IV infusion may substantially increase convenience of treatment. In this review, we outline evidence emerging from clinical trials that shows increasing clinical applicability of Isa across the MM therapeutic spectrum. We then review recent study results demonstrating that new treatment modalities, either SC Isa administration via an on-body delivery system (OBDS) or fast, 30-minute, fixed-volume IV infusion, are safe and effective, and enhance convenience of treatment with Isa for MM patients and healthcare providers. In the recently reported Phase 1b study, the safety profile and efficacy of Isa administered SC plus pomalidomide-dexamethasone were comparable to those observed with Isa administered IV plus pomalidomide-dexamethasone in the control arm and in the ICARIA-MM trial. Analysis of patient-reported outcomes indicated patient confidence in SC Isa administration and satisfaction with treatment delivery by OBDS. These findings point to SC administration as the preferred route for future treatment with Isa-based combinations, as well as to the use of fast, 30-minute IV infusions in settings where SC administration of Isa might not be available.


Sujet(s)
Anticorps monoclonaux humanisés , Myélome multiple , Humains , Myélome multiple/traitement médicamenteux , Perfusions veineuses , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/administration et posologie , Injections sous-cutanées , Antigènes CD38/antagonistes et inhibiteurs
7.
Blood Adv ; 8(11): 2622-2634, 2024 Jun 11.
Article de Anglais | MEDLINE | ID: mdl-38507742

RÉSUMÉ

ABSTRACT: Autoimmune hemolytic anemia (AIHA) is a rare autoantibody-mediated disease. For steroid and/or rituximab-refractory AIHA, there is no consensus on optimal treatment. Daratumumab, a monoclonal antibody targeting CD38, could be beneficial by suppression of CD38+ plasma cells and thus autoantibody secretion. In addition, because CD38 is also expressed by activated T cells, daratumumab may also act via immunomodulatory effects. We evaluated the efficacy and safety of daratumumab monotherapy in an international retrospective study including 19 adult patients with heavily pretreated refractory AIHA. In warm AIHA (wAIHA, n = 12), overall response was 50% with a median response duration of 5.5 months (range, 2-12), including ongoing response in 2 patients after 6 and 12 months. Of 6 nonresponders, 4 had Evans syndrome. In cold AIHA (cAIHA, n = 7) overall hemoglobin (Hb) response was 57%, with ongoing response in 3 of 7 patients. One additional patient with nonanemic cAIHA was treated for severe acrocyanosis and reached a clinical acrocyanosis response as well as a Hb increase. Of 6 patients with cAIHA with acrocyanosis, 4 had improved symptoms after daratumumab treatment. In 2 patients with wAIHA treated with daratumumab, in whom we prospectively collected blood samples, we found complete CD38+ T-cell depletion after daratumumab, as well as altered T-cell subset differentiation and a severely diminished capacity for cell activation and proliferation. Reappearance of CD38+ T cells coincided with disease relapse in 1 patient. In conclusion, our data show that daratumumab therapy may be a treatment option for refractory AIHA. The observed immunomodulatory effects that may contribute to the clinical response deserve further exploration.


Sujet(s)
Anémie hémolytique auto-immune , Anticorps monoclonaux , Humains , Anémie hémolytique auto-immune/traitement médicamenteux , Anticorps monoclonaux/usage thérapeutique , Femelle , Mâle , Adulte d'âge moyen , Adulte , Sujet âgé , Études rétrospectives , Résultat thérapeutique , Antigènes CD38/antagonistes et inhibiteurs
8.
J Neurol Neurosurg Psychiatry ; 95(7): 609-611, 2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38341197

RÉSUMÉ

BACKGROUND: Intravenous immunoglobulin (IVIG) and rituximab are considered the first-line and second-line treatments for Chronic Ataxic Neuropathy and Ophthalmoplegia with IgM-paraprotein, cold Agglutinins, and anti-Disialosyl antibodies (CANOMAD), with an overall clinical response around 50%. New anti-CD38 daratumumab, targeting long-lived plasma cells, has been reported as a promising therapy for treatment-refractory antibody-mediated disorders. We report the first case of a severe refractory CANOMAD, successfully treated with daratumumab. METHODS: A patient in their 70s with severe relapsing CANOMAD, refractory to IVIG, steroids, rituximab and ibrutinib developed severe tetraparesis and respiratory failure. Plasma exchange (PE) improved motor and ventilatory function; however, after 6 weeks, patient remained PE dependent. Intravenous daratumumab was initiated at 16 mg/kg weekly for 3 weeks, every 2 weeks for the second and third month, and monthly afterwards. RESULTS: After 3 weeks of starting daratumumab, PE was discontinued and, since then, the patient evolved to complete recovery. Antidisialosyl antibody titres decreased after PE and remained stable during daratumumab. Serum neurofilament light-chain levels were elevated in the exacerbation phase and normalised after daratumumab. The patient remains in clinical remission under monthly daratumumab, 12 months after initiation. CONCLUSIONS: The first patient with aggressive treatment-refractory CANOMAD treated with daratumumab provides proof-of-principle evidence that daratumumab may be an effective treatment in IgM-related neuropathies.


Sujet(s)
Antigènes CD38 , Anticorps monoclonaux , Humains , Anticorps monoclonaux/usage thérapeutique , Antigènes CD38/antagonistes et inhibiteurs , Sujet âgé , Mâle , Résultat thérapeutique , Échange plasmatique , Ophtalmoplégie/traitement médicamenteux
9.
Clin Lymphoma Myeloma Leuk ; 23 Suppl 2: S248, 2023 Sep.
Article de Anglais | MEDLINE | ID: mdl-38972691

RÉSUMÉ

This article has been retracted: please see Elsevier Policy on Article Withdrawal (https://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the authors. This abstract was submitted to the International Myeloma Society and the meeting organizers by the first author without informed consent of all co-authors.


Sujet(s)
Antigènes CD38 , Myélome multiple , Myélome multiple/traitement médicamenteux , Humains , Antigènes CD38/métabolisme , Antigènes CD38/antagonistes et inhibiteurs , Thérapie moléculaire ciblée/méthodes , Glycoprotéines membranaires
10.
J Med Chem ; 65(13): 9418-9446, 2022 07 14.
Article de Anglais | MEDLINE | ID: mdl-35762533

RÉSUMÉ

CD38 is one of the major nicotinamide adenine dinucleotide (NAD+)- and nicotinamide adenine dinucleotide phosphate (NADP+)-consuming enzymes in mammals. NAD+, NADP+, and their reduced counterparts are essential coenzymes for numerous enzymatic reactions, including the maintenance of cellular and mitochondrial redox balance. CD38 expression is upregulated in age-associated inflammation as well as numerous metabolic diseases, resulting in cellular and mitochondrial dysfunction. Recent literature studies demonstrate that CD38 is activated upon ischemia/reperfusion (I/R), leading to a depletion of NADP+, which results in endothelial damage and myocardial infarction in the heart. Despite increasing evidence of CD38 involvement in various disease states, relatively few CD38 enzymatic inhibitors have been reported to date. Herein, we describe a CD38 enzymatic inhibitor (MK-0159, IC50 = 3 nM against murine CD38) that inhibits CD38 in in vitro assay. Mice treated with MK-0159 show strong protection from myocardial damage upon cardiac I/R injury compared to those treated with NAD+ precursors (nicotinamide riboside) or the known CD38 inhibitor, 78c.


Sujet(s)
Antigènes CD38/antagonistes et inhibiteurs , Glycoprotéines membranaires/antagonistes et inhibiteurs , NAD , Lésion d'ischémie-reperfusion , Animaux , Antienzymes , Ischémie , Mammifères/métabolisme , Souris , NAD/métabolisme , NADP/métabolisme , Lésion d'ischémie-reperfusion/traitement médicamenteux , Lésion d'ischémie-reperfusion/prévention et contrôle
11.
Blood ; 139(8): 1160-1176, 2022 02 24.
Article de Anglais | MEDLINE | ID: mdl-35201323

RÉSUMÉ

Anti-CD38 monoclonal antibodies (mAbs) represent a breakthrough in the treatment of multiple myeloma (MM), yet some patients fail to respond or progress quickly with this therapy, highlighting the need for novel approaches. In this study we compared the preclinical efficacy of SAR442085, a next-generation anti-CD38 mAb with enhanced affinity for activating Fcγ receptors (FcγR), with first-generation anti-CD38 mAb daratumumab and isatuximab. In surface plasmon resonance and cellular binding assays, we found that SAR442085 had higher binding affinity than daratumumab and isatuximab for FcγRIIa (CD32a) and FcγRIIIa (CD16a). SAR442085 also exhibited better in vitro antibody-dependent cellular cytotoxicity (ADCC) against a panel of MM cells expressing variable CD38 receptor densities including MM patients' primary plasma cells. The enhanced ADCC of SAR442085 was confirmed using NK-92 cells bearing low and high affinity FcγRIIIa (CD16a)-158F/V variants. Using MM patients' primary bone marrow cells, we confirmed that SAR442085 had an increased ability to engage FcγRIIIa, resulting in higher natural killer (NK) cell activation and degranulation against primary plasma cells than preexisting Fc wild-type anti-CD38 mAbs. Finally, using huFcgR transgenic mice that express human Fcγ receptors under the control of their human regulatory elements, we demonstrated that SAR442085 had higher NK cell-dependent in vivo antitumor efficacy and better survival than daratumumab and isatuximab against EL4 thymoma or VK*MYC myeloma cells overexpressing human CD38. These results highlight the preclinical efficacy of SAR442085 and support the current evaluation of this next-generation anti-CD38 antibody in phase I clinical development in patients with relapsed/refractory MM.


Sujet(s)
Antigènes CD38/antagonistes et inhibiteurs , Antinéoplasiques immunologiques/pharmacologie , Cellules de la moelle osseuse , Glycoprotéines membranaires/antagonistes et inhibiteurs , Myélome multiple , Protéines tumorales/antagonistes et inhibiteurs , Antigènes CD38/métabolisme , Animaux , Cellules de la moelle osseuse/métabolisme , Cellules de la moelle osseuse/anatomopathologie , Lignée cellulaire tumorale , Cellules HEK293 , Humains , Glycoprotéines membranaires/métabolisme , Souris transgéniques , Myélome multiple/traitement médicamenteux , Myélome multiple/métabolisme , Myélome multiple/anatomopathologie , Protéines tumorales/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
12.
Hematology ; 27(1): 204-207, 2022 Dec.
Article de Anglais | MEDLINE | ID: mdl-35134321

RÉSUMÉ

Objectives: Daratumumab is the first anti-CD38 monoclonal antibody (Mab) used to treat myeloma in the newly diagnosed setting and in the relapsed setting. Isatuximab, another Mab targeting a specific epitope on the CD38 receptor, was recently approved in the UK in combination with pomalidomide and dexamethasone (IsaPomDex) to treat myeloma patients who received three prior lines of therapy. However, there is a lack of understanding of whether using a prior anti-CD38 Mab (e.g. daratumumab) can affect the efficacy of another Mab (e.g. isatuximab), when the latter is used to treat a subsequent relapse.Methods: We performed a UK-wide outcomes study of IsaPomDex in the real-world. In this case series, we report a detailed descriptive analysis of the characteristics and clinical outcomes of five IsaPomDex patients in UK routine practice (Patients I to V), with a prior exposure to daratumumab.Results: Age range was 51-77 years with two patients >70 and three patients <70 years. The cytogenetic risk was standard in two patients, high in two patients and not known in one patient. Prior daratumumab regimen were monotherapy (dara-mono) in one patient (II), and daratumumab with bortezomib and dexamethasone (DVd) in four patients. Responses to prior daratumumab were: very good partial response (VGPR) in two patients (I and III), minor response-stable disease (MR-SD) in one patient (II), and progressive disease (PD) in two patients (IV and V). Median (range) number of IsaPomDex cycles received was 2 (1-4). Outcomes of IsaPomDex were PD in three patients (II, IV and V) and a response in two patients. Response categories were: MR-SD in patient I and PR in patient III.Discussion: Despite the limitations of our case series, we described the first UK real-world report of IsaPomDex outcomes in myeloma patients with a prior exposure to daratumumab.Conclusion: Large prospective studies are required to further evaluate myeloma outcomes in this setting.


Sujet(s)
Anticorps monoclonaux humanisés/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Dexaméthasone/usage thérapeutique , Myélome multiple/traitement médicamenteux , Récidive tumorale locale/traitement médicamenteux , Thalidomide/analogues et dérivés , Antigènes CD38/antagonistes et inhibiteurs , Sujet âgé , Anticorps monoclonaux/usage thérapeutique , Femelle , Humains , Mâle , Glycoprotéines membranaires/antagonistes et inhibiteurs , Adulte d'âge moyen , Thalidomide/usage thérapeutique , Résultat thérapeutique
14.
J Nanobiotechnology ; 19(1): 434, 2021 Dec 20.
Article de Anglais | MEDLINE | ID: mdl-34930285

RÉSUMÉ

BACKGROUND: As an efficient tumor immunotherapy, PD-1 antibody has been gradually used in clinical tumor treatment, but the low response rate and excessive immune response limit its extensive application. RESULTS: Herein, a therapeutic regime for the reinvigoration and activation of the tumor immune microenvironment is introduced to improve the anti-tumor effect of the PD-1 antibody. To comprehensively improve the effect of the immunotherapy and reduce excessive immune response, a biomimetic cascade targeting nanosystem, siRNA@PLOV, which was fused by photothermal sensitive liposomes (PTSLs) and attenuated Salmonella outer membrane vesicles (OMVs), was administered in the tumor therapy for targeting of tumor tissues and T cells within tumor respectively. The fused PLOVs which not only retained the biological character of the OMVs, but also enhanced the drug loading ability. The results demonstrated that the immunogenicity of OMVs and photothermal effects can obviously increase the infiltration of T cells and the silencing of CD38 can effectively improve the T cell cytotoxicity, especially combining with PD-1 antibody. CONCLUSIONS: Interesting, this study revealed that anti-PD-1 administration on the 5th day after siRNA@PLOV treatment had the best performance in killing tumors compared with other groups. In addition, this new therapeutic regime also presents a novel strategy for inducing "vaccine effects", conclusively highlighting its potential in preventing tumor recurrence and improving prognosis.


Sujet(s)
Immunothérapie/méthodes , Tumeurs/thérapie , Vésicules de sécrétion/composition chimique , Antigènes CD38/antagonistes et inhibiteurs , Antigènes CD38/génétique , Antigènes CD38/métabolisme , Animaux , Anticorps monoclonaux/composition chimique , Anticorps monoclonaux/usage thérapeutique , Membrane bactérienne externe/métabolisme , Lignée cellulaire tumorale , Humains , Liposomes/composition chimique , Souris , Souris de lignée BALB C , Souris de lignée ICR , Tumeurs/traitement médicamenteux , Récepteur-1 de mort cellulaire programmée/immunologie , Petit ARN interférent/composition chimique , Petit ARN interférent/usage thérapeutique , Salmonella/métabolisme , Lymphocytes T/cytologie , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Transplantation hétérologue
15.
Molecules ; 26(23)2021 Nov 26.
Article de Anglais | MEDLINE | ID: mdl-34885748

RÉSUMÉ

Although a monoclonal antibody targeting the multifunctional ectoenzyme CD38 is an FDA-approved drug, few small molecule inhibitors exist for this enzyme that catalyzes inter alia the formation and metabolism of the N1-ribosylated, Ca2+-mobilizing, second messenger cyclic adenosine 5'-diphosphoribose (cADPR). N1-Inosine 5'-monophosphate (N1-IMP) is a fragment directly related to cADPR. 8-Substituted-N1-IMP derivatives, prepared by degradation of cyclic parent compounds, inhibit CD38-mediated cADPR hydrolysis more efficiently than related cyclic analogues, making them attractive for inhibitor development. We report a total synthesis of the N1-IMP scaffold from adenine and a small initial compound series that facilitated early delineation of structure-activity parameters, with analogues evaluated for inhibition of CD38-mediated hydrolysis of cADPR. The 5'-phosphate group proved essential for useful activity, but substitution of this group by a sulfonamide bioisostere was not fruitful. 8-NH2-N1-IMP is the most potent inhibitor (IC50 = 7.6 µM) and importantly HPLC studies showed this ligand to be cleaved at high CD38 concentrations, confirming its access to the CD38 catalytic machinery and demonstrating the potential of our fragment approach.


Sujet(s)
Antigènes CD38/antagonistes et inhibiteurs , ADP-ribose cyclique/métabolisme , Inosine/métabolisme , Bibliothèques de petites molécules/composition chimique , Antigènes CD38/métabolisme , Adénosine diphosphate ribose/métabolisme , Calcium/métabolisme , Catalyse/effets des médicaments et des substances chimiques , Humains , Hydrolyse/effets des médicaments et des substances chimiques , IMP/composition chimique , Bibliothèques de petites molécules/pharmacologie , Relation structure-activité
16.
Sci Rep ; 11(1): 22062, 2021 11 11.
Article de Anglais | MEDLINE | ID: mdl-34764342

RÉSUMÉ

Targeting the interaction between leukemic cells and the microenvironment is an appealing approach to enhance the therapeutic efficacy in acute myeloid leukemia (AML). AML infiltration induces a significant release of inflammatory cytokines in the human bone marrow niche which accelerates leukemogenesis. As the transmembrane glycoprotein CD38 has been shown to regulate cytokine release, we assessed the anti-leukemic potential of CD38 inhibition in AML. CD38 expression in AML cells proved to depend on microenvironmental cues and could be significantly enforced through addition of tretinoin. In fact, the anti-CD38 antibody daratumumab showed significant cytostatic efficacy in a 3D in vitro triple-culture model of AML, but with modest cell-autonomous cytotoxic activity and independent of CD38 expression level. In line with a predominantly microenvironment-mediated activity of daratumumab in AML, CD38 inhibition significantly induced antibody-dependent phagocytosis and showed interference with AML cell trafficking in vivo in a xenograft transplantation model, but overall lacked robust anti-leukemic effects.


Sujet(s)
Antigènes CD38/antagonistes et inhibiteurs , Anticorps monoclonaux/usage thérapeutique , Antinéoplasiques immunologiques/usage thérapeutique , Leucémie aigüe myéloïde/traitement médicamenteux , Phagocytose/effets des médicaments et des substances chimiques , Antigènes CD38/immunologie , Animaux , Anticorps monoclonaux/pharmacologie , Antinéoplasiques immunologiques/pharmacologie , Mouvement cellulaire/effets des médicaments et des substances chimiques , Humains , Leucémie aigüe myéloïde/immunologie , Souris de lignée NOD , Cellules cancéreuses en culture , Microenvironnement tumoral/effets des médicaments et des substances chimiques
18.
J Hematol Oncol ; 14(1): 161, 2021 10 09.
Article de Anglais | MEDLINE | ID: mdl-34627333

RÉSUMÉ

BACKGROUND: BCMA-specific chimeric antigen receptor-T cells (CAR-Ts) have exhibited remarkable efficacy in refractory or relapsed multiple myeloma (RRMM); however, primary resistance and relapse exist with single-target immunotherapy. Bispecific CARs are proposed to mitigate these limitations. METHODS: We constructed a humanized bispecific BM38 CAR targeting BCMA and CD38 and tested the antimyeloma activity of BM38 CAR-Ts in vitro and in vivo. Twenty-three patients with RRMM received infusions of BM38 CAR-Ts in a phase I trial. RESULTS: BM38 CAR-Ts showed stronger in vitro cytotoxicity to heterogeneous MM cells than did T cells expressing an individual BCMA or CD38 CAR. BM38 CAR-Ts also exhibited potent antimyeloma activity in xenograft mouse models. In the phase I trial, cytokine release syndrome occurred in 20 patients (87%) and was mostly grade 1-2 (65%). Neurotoxicity was not observed. Hematologic toxicities were common, including neutropenia in 96% of the patients, leukopenia in 87%, anemia in 43% and thrombocytopenia in 61%. At a median follow-up of 9.0 months (range 0.5 to 18.5), 20 patients (87%) attained a clinical response and minimal residual disease-negativity (≤ 10-4 nucleated cells), with 12 (52%) achieving a stringent complete response. Extramedullary plasmacytoma was eliminated completely in 56% and partially in 33% and of 9 patients. The median progression-free survival was 17.2 months. Two relapsed patients maintained BCMA and CD38 expression on MM cells. Notably, BM38 CAR-Ts cells were detectable in 77.8% of evaluable patients at 9 months and 62.2% at 12 months. CONCLUSION: Bispecific BM38 CAR-Ts were feasible, safe and significantly effective in patient with RRMM. TRIAL REGISTRATION: Chictr.org.cn ChiCTR1800018143.


Sujet(s)
Antigènes CD38/immunologie , Antigène de maturation des cellules B/immunologie , Immunothérapie adoptive/méthodes , Myélome multiple/thérapie , Récepteurs chimériques pour l'antigène/usage thérapeutique , Antigènes CD38/antagonistes et inhibiteurs , Adulte , Sujet âgé , Animaux , Antigène de maturation des cellules B/antagonistes et inhibiteurs , Lignée cellulaire tumorale , Femelle , Humains , Immunothérapie adoptive/effets indésirables , Mâle , Souris , Adulte d'âge moyen , Simulation de docking moléculaire , Myélome multiple/immunologie , Récidive tumorale locale/immunologie , Récidive tumorale locale/thérapie , Récepteurs chimériques pour l'antigène/immunologie
19.
BMC Cancer ; 21(1): 994, 2021 Sep 06.
Article de Anglais | MEDLINE | ID: mdl-34488679

RÉSUMÉ

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.


Sujet(s)
Antigènes CD38/antagonistes et inhibiteurs , Anticorps monoclonaux/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Antigène CD274/antagonistes et inhibiteurs , Glycoprotéines membranaires/antagonistes et inhibiteurs , Myélome multiple/traitement médicamenteux , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Famille des molécules de signalisation de l'activation des lymphocytes/antagonistes et inhibiteurs , Antigènes CD38/immunologie , Antigène CD274/immunologie , Bortézomib/administration et posologie , Dexaméthasone/administration et posologie , Humains , Facteurs immunologiques/usage thérapeutique , Glycoprotéines membranaires/immunologie , Myélome multiple/immunologie , Myélome multiple/métabolisme , Myélome multiple/anatomopathologie , Prednisone/administration et posologie , Pronostic , Récepteur-1 de mort cellulaire programmée/immunologie , Essais contrôlés randomisés comme sujet , Famille des molécules de signalisation de l'activation des lymphocytes/immunologie
20.
J Neurosci ; 41(41): 8644-8667, 2021 10 13.
Article de Anglais | MEDLINE | ID: mdl-34493542

RÉSUMÉ

Western-style diets cause disruptions in myelinating cells and astrocytes within the mouse CNS. Increased CD38 expression is present in the cuprizone and experimental autoimmune encephalomyelitis models of demyelination and CD38 is the main nicotinamide adenine dinucleotide (NAD+)-depleting enzyme in the CNS. Altered NAD+ metabolism is linked to both high fat consumption and multiple sclerosis (MS). Here, we identify increased CD38 expression in the male mouse spinal cord following chronic high fat consumption, after focal toxin [lysolecithin (LL)]-mediated demyelinating injury, and in reactive astrocytes within active MS lesions. We demonstrate that CD38 catalytically inactive mice are substantially protected from high fat-induced NAD+ depletion, oligodendrocyte loss, oxidative damage, and astrogliosis. A CD38 inhibitor, 78c, increased NAD+ and attenuated neuroinflammatory changes induced by saturated fat applied to astrocyte cultures. Conditioned media from saturated fat-exposed astrocytes applied to oligodendrocyte cultures impaired myelin protein production, suggesting astrocyte-driven indirect mechanisms of oligodendrogliopathy. In cerebellar organotypic slice cultures subject to LL-demyelination, saturated fat impaired signs of remyelination effects that were mitigated by concomitant 78c treatment. Significantly, oral 78c increased counts of oligodendrocytes and remyelinated axons after focal LL-induced spinal cord demyelination. Using a RiboTag approach, we identified a unique in vivo brain astrocyte translatome profile induced by 78c-mediated CD38 inhibition in mice, including decreased expression of proinflammatory astrocyte markers and increased growth factors. Our findings suggest that a high-fat diet impairs oligodendrocyte survival and differentiation through astrocyte-linked mechanisms mediated by the NAD+ase CD38 and highlights CD38 inhibitors as potential therapeutic candidates to improve myelin regeneration.SIGNIFICANCE STATEMENT Myelin disturbances and oligodendrocyte loss can leave axons vulnerable, leading to permanent neurologic deficits. The results of this study suggest that metabolic disturbances, triggered by consumption of a diet high in fat, promote oligodendrogliopathy and impair myelin regeneration through astrocyte-linked indirect nicotinamide adenine dinucleotide (NAD+)-dependent mechanisms. We demonstrate that restoring NAD+ levels via genetic inactivation of CD38 can overcome these effects. Moreover, we show that therapeutic inactivation of CD38 can enhance myelin regeneration. Together, these findings point to a new metabolic targeting strategy positioned to improve disease course in multiple sclerosis and other conditions in which the integrity of myelin is a key concern.


Sujet(s)
Antigènes CD38/métabolisme , Astrocytes/métabolisme , Glycoprotéines membranaires/métabolisme , Gaine de myéline/métabolisme , NAD nucleosidase/physiologie , Régénération nerveuse/physiologie , Remyélinisation/physiologie , Antigènes CD38/antagonistes et inhibiteurs , Antigènes CD38/génétique , Animaux , Cervelet/métabolisme , Alimentation riche en graisse/effets indésirables , Mâle , Glycoprotéines membranaires/antagonistes et inhibiteurs , Glycoprotéines membranaires/génétique , Souris , Souris de lignée C57BL , Souris transgéniques , Gaine de myéline/génétique , Techniques de culture d'organes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...