Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 425
Filter
1.
Front Immunol ; 15: 1406353, 2024.
Article in English | MEDLINE | ID: mdl-38881900

ABSTRACT

An appropriately designed pharmacokinetic (PK) assay that is sensitive for anti-drug antibody (ADA) impact on relevant exposure is an alternative strategy to understand the neutralizing potential of ADAs. However, guidance on how to develop such PK assays and how to confirm the functional ADA impact on exposure is missing. Here, the PK assay of a T-cell-engaging bispecific antibody, cibisatamab, was developed based on its mechanism of action (MoA). Using critical monoclonal anti-idiotypic (anti-ID) antibody positive controls as ADA surrogates, the impact on exposure was evaluated pre-clinically. In a phase I clinical trial (NCT02324257), initial data suggest that the combination of ADA and PK assays for correlation of the ADA response with cibisatamab exposure. To understand the neutralizing potential of patient-derived ADAs on drug activity, advanced ADA characterization has been performed. Structural binding analysis of ADAs to antibody domains of the drug and its impact on targeting were assessed. For this purpose, relevant patient ADA binding features were identified and compared with the specific monoclonal anti-ID antibody-positive controls. Comparable results of target binding inhibition and similar impacts on exposure suggest that the observed reduction of Cmax and Ctrough levels in patients is caused by the neutralizing potential of ADAs and allows a correlation between ADA response and loss of exposure. Therefore, the described study provides important functional aspects for the development of an appropriately designed PK assay for bispecific antibodies as an alternative option towards understanding the neutralizing ADA impact on exposure.


Subject(s)
Antibodies, Bispecific , T-Lymphocytes , Humans , Antibodies, Bispecific/immunology , Antibodies, Bispecific/pharmacology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Antibodies, Anti-Idiotypic/immunology , Antibodies, Neutralizing/immunology
2.
Biochimie ; 2024 Jun 11.
Article in English | MEDLINE | ID: mdl-38871044

ABSTRACT

Proteases are key enzymes in viral replication, and interfering with these targets is the basis for therapeutic interventions. We previously introduced a hypothesis about conformational selection in the protease of dengue virus and related flaviviruses, based on conformational plasticity noted in X-ray structures. The present work presents the first functional evidence for alternate recognition by the dengue protease, in a mechanism based primarily on conformational selection rather than induced-fit. Recognition of distinct substrates and inhibitors in proteolytic and binding assays varies to a different extent, depending on factors reported to influence the protease structure. The pH, salinity, buffer type, and temperature cause a change in binding, proteolysis, or inhibition behavior. Using representative inhibitors with distinct structural scaffolds, we identify two contrasting binding profiles to dengue protease. Noticeable effects are observed in the binding assay upon inclusion of a non-ionic detergent in comparison to the proteolytic assay. The findings highlight the impact of the selection of testing conditions on the observed ligand affinity or inhibitory potency. From a broader scope, the dengue protease presents an example, where the induced-fit paradigm appears insufficient to explain binding events with the biological target. Furthermore, this protein reveals the complexity of comparing or combining biochemical assay data obtained under different conditions. This can be particularly critical for artificial intelligence (AI) approaches in drug discovery that rely on large datasets of compounds activity, compiled from different sources using non-identical testing procedures. In such cases, mismatched results will compromise the model quality and its predictive power.

3.
Front Immunol ; 15: 1401471, 2024.
Article in English | MEDLINE | ID: mdl-38938560

ABSTRACT

TRIM21 is a pivotal effector in the immune system, orchestrating antibody-mediated responses and modulating immune signaling. In this comprehensive study, we focus on the interaction of TRIM21 with Fc engineered antibodies and subsequent implications for viral neutralization. Through a series of analytical techniques, including biosensor assays, mass photometry, and electron microscopy, along with structure predictions, we unravel the intricate mechanisms governing the interplay between TRIM21 and antibodies. Our investigations reveal that the TRIM21 capacity to recognize, bind, and facilitate the proteasomal degradation of antibody-coated viruses is critically dependent on the affinity and avidity interplay of its interactions with antibody Fc regions. We suggest a novel binding mechanism, where TRIM21 binding to one Fc site results in the detachment of PRYSPRY from the coiled-coil domain, enhancing mobility due to its flexible linker, thereby facilitating the engagement of the second site, resulting in avidity due to bivalent engagement. These findings shed light on the dual role of TRIM21 in antiviral immunity, both in recognizing and directing viruses for intracellular degradation, and demonstrate its potential for therapeutic exploitation. The study advances our understanding of intracellular immune responses and opens new avenues for the development of antiviral strategies and innovation in tailored effector functions designed to leverage TRIM21s unique binding mode.


Subject(s)
Antibodies, Neutralizing , Immunoglobulin Fc Fragments , Protein Binding , Ribonucleoproteins , Humans , Ribonucleoproteins/immunology , Ribonucleoproteins/metabolism , Antibodies, Neutralizing/immunology , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fc Fragments/metabolism , Protein Engineering , Antibodies, Viral/immunology , Antibodies, Viral/metabolism , Antibody Affinity/immunology , Animals
4.
Adv Sci (Weinh) ; : e2400640, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38810019

ABSTRACT

Organoboron compounds have a wide range of applications in numerous research fields, and metmhods to incorporate them in biomolecules are much sought after. Here, on-resin chemical syntheses of aliphatic and vinylogous peptide boronic acids are presented by transition metal-catalyzed late-stage hydroboration of alkene and alkyne groups in peptides and peptoids, for example on allyl- and propargylglycine residues, using readily available chemicals. These methods yield peptide boronic acids with much shorter linkers than previously reported on-resin methods. Furthermore, the methods are regio- and stereoselective, compatible with all canonical amino acid residues and can be applied to short, long, and in part even "difficult" peptide sequences. In a feasibility study, the protected peptide vinylboronic acids are further derivatized by the Petasis reaction using salicylaldehyde derivatives. The ability of the obtained peptide boronic acids to reversibly bind to carbohydrates is demonstrated in a catch-release model experiment using a fluorescently labeled peptide boronic acid on cross-linked dextran beads. In summary, this highlights the potential of the target compounds for drug discovery, glycan-specific target recognition, controlled release, and diagnostics.

5.
Front Oncol ; 14: 1346502, 2024.
Article in English | MEDLINE | ID: mdl-38577337

ABSTRACT

Introduction: Although checkpoint inhibitors (CPIs) have improved outcomes for patients with metastatic melanoma, those progressing on CPIs have limited therapeutic options. To address this unmet need and overcome CPI resistance mechanisms, novel immunotherapies, such as T-cell engaging agents, are being developed. The use of these agents has sometimes been limited by the immune response mounted against them in the form of anti-drug antibodies (ADAs), which is challenging to predict preclinically and can lead to neutralization of the drug and loss of efficacy. Methods: TYRP1-TCB (RO7293583; RG6232) is a T-cell engaging bispecific (TCB) antibody that targets tyrosinase-related protein 1 (TYRP1), which is expressed in many melanomas, thereby directing T cells to kill TYRP1-expressing tumor cells. Preclinical studies show TYRP1-TCB to have potent anti-tumor activity. This first-in-human (FIH) phase 1 dose-escalation study characterized the safety, tolerability, maximum tolerated dose/optimal biological dose, and pharmacokinetics (PK) of TYRP1-TCB in patients with metastatic melanoma (NCT04551352). Results: Twenty participants with cutaneous, uveal, or mucosal TYRP1-positive melanoma received TYRP1-TCB in escalating doses (0.045 to 0.4 mg). All participants experienced ≥1 treatment-related adverse event (TRAE); two participants experienced grade 3 TRAEs. The most common toxicities were grade 1-2 cytokine release syndrome (CRS) and rash. Fractionated dosing mitigated CRS and was associated with lower levels of interleukin-6 and tumor necrosis factor-alpha. Measurement of active drug (dual TYPR1- and CD3-binding) PK rapidly identified loss of active drug exposure in all participants treated with 0.4 mg in a flat dosing schedule for ≥3 cycles. Loss of exposure was associated with development of ADAs towards both the TYRP1 and CD3 domains. A total drug PK assay, measuring free and ADA-bound forms, demonstrated that TYRP1-TCB-ADA immune complexes were present in participant samples, but showed no drug activity in vitro. Discussion: This study provides important insights into how the use of active drug PK assays, coupled with mechanistic follow-up, can inform and enable ongoing benefit/risk assessment for individuals participating in FIH dose-escalation trials. Translational studies that lead to a better understanding of the underlying biology of cognate T- and B-cell interactions, ultimately resulting in ADA development to novel biotherapeutics, are needed.

6.
Br J Sports Med ; 58(13): 709-716, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38684328

ABSTRACT

BACKGROUND: To identify mechanisms and patterns of anterior cruciate ligament (ACL) injury in adult women's professional football by means of video match analysis. METHODS: ACL match injuries sustained in Germany's first women's league during the 2016-2017 to 2022-2023 seasons were prospectively analysed by three expert raters using a standardised observation form. Epidemiological and injury data, as well as the medical history of ACL tears, were obtained from media reports and the statutory accident insurance for professional athletes. RESULTS: Thirty-seven ACL injuries sustained in official football matches were included in the video analysis, of which 24 (65%) had associated knee injuries, mainly meniscus and collateral ligament injuries. According to the categorised contact mechanisms, 17 (46%) were non-contact injuries, 14 indirect contact injuries (38%) and six direct contact injuries (16%). Of the 17 non-contact injuries, seven (41%) occurred during the first 15 min of the match. Contact mechanisms did not differ between primary and secondary ACL injuries to the same or the contralateral side. Most injuries (80%) of field players occurred during horizontal movements such as sprinting (n=9, 26%), change-of-direction manoeuvres (n=7, 19%), stopping (n=5, 14%) and lunging (n=5, 14%). Four distinct repetitive patterns of ACL match injuries were identified: (1) non-contact 'pressing ACL injury' (n=9), (2) indirect contact 'parallel sprinting and tackling ACL injury' (n=7), (3) direct contact 'knee-to-knee ACL injury' (n=6) and (4) non-contact 'landing ACL injury' (n=4). CONCLUSION: Most of the identified patterns of ACL injuries in women's professional football have great potential for prevention.


Subject(s)
Anterior Cruciate Ligament Injuries , Soccer , Video Recording , Humans , Anterior Cruciate Ligament Injuries/epidemiology , Soccer/injuries , Female , Adult , Prospective Studies , Germany/epidemiology , Young Adult , Athletic Injuries/epidemiology
7.
Orthopadie (Heidelb) ; 53(6): 420-426, 2024 Jun.
Article in German | MEDLINE | ID: mdl-38662137

ABSTRACT

High injury rates and long injury-related downtimes demonstrate the need for effective and differentiated injury prevention strategies in football. Preventive measures should take place in various fields and should not be reduced to training programmes or medical approaches. The so-called "Big 6 of injury prevention" provide an overview of the key areas that should be addressed. In modern sports medicine, it should be a matter of course that the team doctor already participates in the primary prevention strategies and does not only get involved in the post-traumatic treatment. Similarly, a decision on a player's return to play after an injury should not be based solely on the medical assessment of the team doctor. Good communication and interdisciplinary cooperation, therefore, form the basis for successful prevention.


Subject(s)
Athletic Injuries , Soccer , Humans , Athletic Injuries/prevention & control , Mentoring/methods , Soccer/injuries , Sports Medicine
8.
Clin Exp Immunol ; 217(1): 15-30, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38642912

ABSTRACT

B and T cells collaborate to drive autoimmune disease (AID). Historically, B- and T-cell (B-T cell) co-interaction was targeted through different pathways such as alemtuzumab, abatacept, and dapirolizumab with variable impact on B-cell depletion (BCD), whereas the majority of patients with AID including rheumatoid arthritis, systemic lupus erythematosus, multiple sclerosis, and organ transplantation benefit from targeted BCD with anti-CD20 monoclonal antibodies such as rituximab, ocrelizumab, or ofatumumab. Refractory AID is a significant problem for patients with incomplete BCD with a greater frequency of IgD-CD27+ switched memory B cells, CD19+CD20- B cells, and plasma cells that are not directly targeted by anti-CD20 antibodies, whereas most lymphoid tissue plasma cells express CD19. Furthermore, B-T-cell collaboration is predominant in lymphoid tissues and at sites of inflammation such as the joint and kidney, where BCD may be inefficient, due to limited access to key effector cells. In the treatment of cancer, chimeric antigen receptor (CAR) T-cell therapy and T-cell engagers (TCE) that recruit T cells to induce B-cell cytotoxicity have delivered promising results for anti-CD19 CAR T-cell therapies, the CD19 TCE blinatumomab and CD20 TCE such as mosunetuzumab, glofitamab, or epcoritamab. Limited evidence suggests that anti-CD19 CAR T-cell therapy may be effective in managing refractory AID whereas we await evaluation of TCE for use in non-oncological indications. Therefore, here, we discuss the potential mechanistic advantages of novel therapies that rely on T cells as effector cells to disrupt B-T-cell collaboration toward overcoming rituximab-resistant AID.


Subject(s)
Autoimmune Diseases , B-Lymphocytes , Immunotherapy, Adoptive , T-Lymphocytes , Humans , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , B-Lymphocytes/immunology , T-Lymphocytes/immunology , Immunotherapy, Adoptive/methods , Receptors, Chimeric Antigen/immunology , Rituximab/therapeutic use , Cell Communication/immunology , Animals
9.
Nat Commun ; 15(1): 3271, 2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38627373

ABSTRACT

Selective binding of TCR-like antibodies that target a single tumour-specific peptide antigen presented by human leukocyte antigens (HLA) is the absolute prerequisite for their therapeutic suitability and patient safety. To date, selectivity assessment has been limited to peptide library screening and predictive modeling. We developed an experimental platform to de novo identify interactomes of TCR-like antibodies directly in human tissues using mass spectrometry. As proof of concept, we confirm the target epitope of a MAGE-A4-specific TCR-like antibody. We further determine cross-reactive peptide sequences for ESK1, a TCR-like antibody with known off-target activity, in human liver tissue. We confirm off-target-induced T cell activation and ESK1-mediated liver spheroid killing. Off-target sequences feature an amino acid motif that allows a structural groove-coordination mimicking that of the target peptide, therefore allowing the interaction with the engager molecule. We conclude that our strategy offers an accurate, scalable route for evaluating the non-clinical safety profile of TCR-like antibody therapeutics prior to first-in-human clinical application.


Subject(s)
Antibodies , Peptides , Humans , Cell Line, Tumor , Peptides/chemistry , Antigens, Neoplasm , Receptors, Antigen, T-Cell/metabolism
10.
Nat Rev Drug Discov ; 23(4): 301-319, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38448606

ABSTRACT

Bispecific antibodies (bsAbs) enable novel mechanisms of action and/or therapeutic applications that cannot be achieved using conventional IgG-based antibodies. Consequently, development of these molecules has garnered substantial interest in the past decade and, as of the end of 2023, 14 bsAbs have been approved: 11 for the treatment of cancer and 3 for non-oncology indications. bsAbs are available in different formats, address different targets and mediate anticancer function via different molecular mechanisms. Here, we provide an overview of recent developments in the field of bsAbs for cancer therapy. We focus on bsAbs that are approved or in clinical development, including bsAb-mediated dual modulators of signalling pathways, tumour-targeted receptor agonists, bsAb-drug conjugates, bispecific T cell, natural killer cell and innate immune cell engagers, and bispecific checkpoint inhibitors and co-stimulators. Finally, we provide an outlook into next-generation bsAbs in earlier stages of development, including trispecifics, bsAb prodrugs, bsAbs that induce degradation of tumour targets and bsAbs acting as cytokine mimetics.


Subject(s)
Antibodies, Bispecific , Neoplasms , Humans , Signal Transduction
11.
J Clin Periodontol ; 51(7): 915-925, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38508585

ABSTRACT

AIM: To evaluate the suitability of a Doppler ultrasound probe in detecting the greater palatine artery or its greater branches non-invasively. MATERIALS AND METHODS: The palatal mucosa of 108 participants (median age 34 years, 51 female) was systematically divided into transversal sectors, each aligning with the positions of the upper molars (M), premolars (P) and canine teeth (C), aiming to facilitate precise and consistent localization of the detected palatal blood vessel across different patients. Blood flow of the palatal blood vessels, presumably, was located by scanning the palatal vault bilaterally using an 8-MHz ultrasound probe linked to a transducer. The distance to the corresponding tooth was measured using a millimetre-scale periodontal probe. RESULTS: Within the regions of M2 to P1, the ultrasound transducer gave a delimitable acoustic pulse signal in 80%-98% of all measurements. The measured median distances between the determined position of the artery and the corresponding teeth ranged from 13 to 15 mm, with smaller distances in the anterior region. In several sectors, the distance was significantly higher for men (C: p = .048; P1: p = .041, M1: p < .01; M2: p = .034). CONCLUSIONS: Use of the Doppler ultrasound transducer might be a promising approach to non-invasively detect relevant palatine blood vessels preoperatively. It, therefore, might have the potential to reduce the risk of accidental injury during palatal surgery.


Subject(s)
Palate , Ultrasonography, Doppler , Humans , Cross-Sectional Studies , Male , Female , Adult , Ultrasonography, Doppler/methods , Palate/blood supply , Palate/diagnostic imaging , Middle Aged , Proof of Concept Study , Young Adult
12.
Blood ; 143(21): 2152-2165, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38437725

ABSTRACT

ABSTRACT: Effective T-cell responses not only require the engagement of T-cell receptors (TCRs; "signal 1"), but also the availability of costimulatory signals ("signal 2"). T-cell bispecific antibodies (TCBs) deliver a robust signal 1 by engaging the TCR signaling component CD3ε, while simultaneously binding to tumor antigens. The CD20-TCB glofitamab redirects T cells to CD20-expressing malignant B cells. Although glofitamab exhibits strong single-agent efficacy, adding costimulatory signaling may enhance the depth and durability of T-cell-mediated tumor cell killing. We developed a bispecific CD19-targeted CD28 agonist (CD19-CD28), RG6333, to enhance the efficacy of glofitamab and similar TCBs by delivering signal 2 to tumor-infiltrating T cells. CD19-CD28 distinguishes itself from the superagonistic antibody TGN1412, because its activity requires the simultaneous presence of a TCR signal and CD19 target binding. This is achieved through its engineered format incorporating a mutated Fc region with abolished FcγR and C1q binding, CD28 monovalency, and a moderate CD28 binding affinity. In combination with glofitamab, CD19-CD28 strongly increased T-cell effector functions in ex vivo assays using peripheral blood mononuclear cells and spleen samples derived from patients with lymphoma and enhanced glofitamab-mediated regression of aggressive lymphomas in humanized mice. Notably, the triple combination of glofitamab with CD19-CD28 with the costimulatory 4-1BB agonist, CD19-4-1BBL, offered substantially improved long-term tumor control over glofitamab monotherapy and respective duplet combinations. Our findings highlight CD19-CD28 as a safe and highly efficacious off-the-shelf combination partner for glofitamab, similar TCBs, and other costimulatory agonists. CD19-CD28 is currently in a phase 1 clinical trial in combination with glofitamab. This trial was registered at www.clinicaltrials.gov as #NCT05219513.


Subject(s)
Antibodies, Bispecific , Antigens, CD19 , Antigens, CD20 , CD28 Antigens , Immunotherapy , Humans , CD28 Antigens/immunology , CD28 Antigens/agonists , Animals , Mice , Antibodies, Bispecific/pharmacology , Antigens, CD19/immunology , Antigens, CD20/immunology , Immunotherapy/methods , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays , Mice, Inbred NOD
13.
Sci Transl Med ; 16(737): eadi0295, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38446903

ABSTRACT

Anti-CD20 therapy to deplete B cells is highly efficacious in preventing new white matter lesions in patients with relapsing-remitting multiple sclerosis (RRMS), but its protective capacity against gray matter injury and axonal damage is unclear. In a passive experimental autoimmune encephalomyelitis (EAE) model whereby TH17 cells promote brain leptomeningeal immune cell aggregates, we found that anti-CD20 treatment effectively spared myelin content and prevented myeloid cell activation, oxidative damage, and mitochondrial stress in the subpial gray matter. Anti-CD20 treatment increased B cell survival factor (BAFF) in the serum, cerebrospinal fluid, and leptomeninges of mice with EAE. Although anti-CD20 prevented gray matter demyelination, axonal loss, and neuronal atrophy, co-treatment with anti-BAFF abrogated these benefits. Consistent with the murine studies, we observed that elevated BAFF concentrations after anti-CD20 treatment in patients with RRMS were associated with better clinical outcomes. Moreover, BAFF promoted survival of human neurons in vitro. Together, our data demonstrate that BAFF exerts beneficial functions in MS and EAE in the context of anti-CD20 treatment.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis, Relapsing-Remitting , Humans , Animals , Mice , Neuroprotection , Brain , Gray Matter , Antigen Presentation , Atrophy , Encephalomyelitis, Autoimmune, Experimental/drug therapy
14.
MAbs ; 16(1): 2321635, 2024.
Article in English | MEDLINE | ID: mdl-38465614

ABSTRACT

Bispecific antibodies (bsAbs) are a class of antibodies that can mediate novel mechanisms of action compared to monospecific monoclonal antibodies (mAbs). Since the discovery of mAbs and their adoption as therapeutic agents in the 1980s and 1990s, the development of bsAbs has held substantial appeal. Nevertheless, only three bsAbs (catumaxomab, blinatumomab, emicizumab) were approved through the end of 2020. However, since then, 11 bsAbs received regulatory agency approvals, of which nine (amivantamab, tebentafusp, mosunetuzumab, cadonilimab, teclistamab, glofitamab, epcoritamab, talquetamab, elranatamab) were approved for the treatment of cancer and two (faricimab, ozoralizumab) in non-oncology indications. Notably, of the 13 currently approved bsAbs, two, emicizumab and faricimab, have achieved blockbuster status, showing the promise of this novel class of therapeutics. In the 2020s, the approval of additional bsAbs can be expected in hematological malignancies, solid tumors and non-oncology indications, establishing bsAbs as essential part of the therapeutic armamentarium.


Subject(s)
Antibodies, Bispecific , Antineoplastic Agents , Hematologic Neoplasms , Neoplasms , Humans , Antibodies, Monoclonal/therapeutic use , Hematologic Neoplasms/drug therapy
15.
J Neurochem ; 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38481090

ABSTRACT

Xanthurenic acid (XA) raises a growing multidisciplinary interest based upon its oxidizing properties, its ability to complex certain metal ions, and its detoxifier capacity of 3-hydroxykynurenine (3-HK), its brain precursor. However, little is still known about the role and mechanisms of action of XA in the central nervous system (CNS). Therefore, many research groups have recently investigated XA and its central functions extensively. The present paper critically reviews and discusses all major data related to XA properties and neuronal activities to contribute to the improvement of the current knowledge on XA's central roles and mechanisms of action. In particular, our data showed the existence of a specific G-protein-coupled receptor (GPCR) for XA localized exclusively in brain neurons exhibiting Ca2+ -dependent dendritic release and specific electrophysiological responses. XA properties and central activities suggest a role for this compound in brain intercellular signaling. Indeed, XA stimulates cerebral dopamine (DA) release contrary to its structural analog, kynurenic acid (KYNA). Thus, KYNA/XA ratio could be fundamental in the regulation of brain glutamate and DA release. Cerebral XA may also represent an homeostatic signal between the periphery and several brain regions where XA accumulates easily after peripheral administration. Therefore, XA status in certain psychoses or neurodegenerative diseases seems to be reinforced by its brain-specific properties in balance with its formation and peripheral inputs.

16.
Clin Cancer Res ; 30(8): 1630-1641, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38319672

ABSTRACT

PURPOSE: The immunocytokine cergutuzumab amunaleukin (CEA-IL2v) showed manageable safety and favorable pharmacodynamics in phase I/Ib trials in patients with advanced/metastatic carcinoembryonic antigen-positive (CEA+) solid tumors, but this was accompanied by a high incidence of anti-drug antibodies (ADA). We examined B-cell depletion with obinutuzumab as a potential mitigation strategy. EXPERIMENTAL DESIGN: Preclinical data comparing B-cell depletion with rituximab versus obinutuzumab are summarized. Substudies of phase I/Ib trials investigated the effect of obinutuzumab pretreatment on ADA development, safety, pharmacodynamics, and antitumor activity of CEA-IL2v ± atezolizumab in patients with advanced/metastatic or unresectable CEA+ solid tumors who had progressed on standard of care. RESULTS: Preclinical data showed superior B-cell depletion with obinutuzumab versus rituximab. In clinical studies, patients received CEA-IL2v monotherapy with (n = 16) or without (n = 6) obinutuzumab pretreatment (monotherapy study), or CEA-IL2v + atezolizumab + obinutuzumab pretreatment (n = 5; combination study). In the monotherapy study, after four cycles (every 2 weeks treatment), 0/15 evaluable patients administered obinutuzumab pretreatment had ADAs versus 4/6 patients without obinutuzumab. Obinutuzumab pretreatment with CEA-IL2v monotherapy showed no new safety signals and pharmacodynamic data suggested minimal impact on T cells and natural killer cells. Conversely, increased liver toxicity was observed in the combination study (CEA-IL2v + atezolizumab + obinutuzumab pretreatment). CONCLUSIONS: These preliminary findings suggest that obinutuzumab pretreatment before CEA-IL2v administration in patients with CEA+ solid tumors may be a feasible and potent ADA mitigation strategy, with an acceptable safety profile, supporting broader investigation of obinutuzumab pretreatment for ADA mitigation in other settings.


Subject(s)
Antibodies, Monoclonal, Humanized , Carcinoembryonic Antigen , Neoplasms , Humans , Rituximab , Neoplasms/drug therapy
17.
J Cardiovasc Pharmacol ; 83(1): 126-130, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38180458

ABSTRACT

ABSTRACT: Central sleep apnea (CSA) is common in patients with heart failure. Recent studies link ticagrelor use with CSA. We aimed to evaluate CSA prevalence in patients with coronary heart disease (CHD) and whether ticagrelor use is associated with CSA. We reviewed consecutive patients with CHD who underwent a polysomnography (PSG) test over a 5-year period from 3 sleep centers. We sampled patients who were on ticagrelor or clopidogrel during a PSG test at a 1:4 ticagrelor:clopidogrel ratio. Patients with an active opioid prescription during PSG test were excluded. Age, left ventricle (LV) dysfunction, and P2Y12 inhibitor use were included in a multivariate logistic regression. A total of 135 patients were included with 26 on ticagrelor and 109 on clopidogrel (age 64.1 ± 11.4, 32% male). High CSA burden (12%) and strict CSA (4.4%) were more common in patients on ticagrelor than in those on clopidogrel (27% vs. 8.3% and 10.0% vs. 1.8%). Ticagrelor use (vs. clopidogrel) was associated with high CSA burden (OR 3.53, 95% CI 1.04-12.9, P = 0.039) and trended toward significance for strict CSA (OR 6.32, 95% CI 1.03-51.4, P = 0.052) when adjusting for age and LV dysfunction. In an additional analysis also adjusting for history of atrial fibrillation, ticagrelor use and strict CSA became significantly associated (OR 10.0, 95% CI 1.32-117, P = 0.035). CSA was uncommon in patients with CHD undergoing sleep studies. Ticagrelor use (vs. clopidogrel) was associated with high CSA burden and trended toward significance for strict CSA.


Subject(s)
Coronary Disease , Sleep Apnea, Central , Humans , Male , Middle Aged , Aged , Female , Sleep Apnea, Central/chemically induced , Sleep Apnea, Central/diagnosis , Sleep Apnea, Central/epidemiology , Clopidogrel , Ticagrelor/adverse effects , Analgesics, Opioid , Coronary Disease/diagnosis , Coronary Disease/drug therapy , Coronary Disease/epidemiology
19.
Blood ; 143(1): 57-63, 2024 01 04.
Article in English | MEDLINE | ID: mdl-37824808

ABSTRACT

ABSTRACT: Bruton tyrosine kinase inhibitors (BTKis) that target B-cell receptor signaling have led to a paradigm shift in chronic lymphocytic leukemia (CLL) treatment. BTKis have been shown to reduce abnormally high CLL-associated T-cell counts and the expression of immune checkpoint receptors concomitantly with tumor reduction. However, the impact of BTKi therapy on T-cell function has not been fully characterized. Here, we performed longitudinal immunophenotypic and functional analysis of pretreatment and on-treatment (6 and 12 months) peripheral blood samples from patients in the phase 3 E1912 trial comparing ibrutinib-rituximab with fludarabine, cyclophosphamide, and rituximab (FCR). Intriguingly, we report that despite reduced overall T-cell counts; higher numbers of T cells, including effector CD8+ subsets at baseline and at the 6-month time point, associated with no infections; and favorable progression-free survival in the ibrutinib-rituximab arm. Assays demonstrated enhanced anti-CLL T-cell killing function during ibrutinib-rituximab treatment, including a switch from predominantly CD4+ T-cell:CLL immune synapses at baseline to increased CD8+ lytic synapses on-therapy. Conversely, in the FCR arm, higher T-cell numbers correlated with adverse clinical responses and showed no functional improvement. We further demonstrate the potential of exploiting rejuvenated T-cell cytotoxicity during ibrutinib-rituximab treatment, using the bispecific antibody glofitamab, supporting combination immunotherapy approaches.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Humans , Rituximab , Monitoring, Immunologic , Antineoplastic Combined Chemotherapy Protocols , Cyclophosphamide , Immunotherapy , CD8-Positive T-Lymphocytes
20.
Front Cell Dev Biol ; 11: 1270398, 2023.
Article in English | MEDLINE | ID: mdl-38020903

ABSTRACT

Introduction: The glycoengineered type II anti-CD20 monoclonal antibody obinutuzumab has been licensed for treatment in follicular non-Hodgkin lymphoma and B-CLL following clinical trials demonstrating superior outcomes to standard of care treatment. However, ultimately many patients still relapse, highlighting the need to understand the mechanisms behind treatment failure to improve patient care. Resistance to chemotherapy is often caused by the ability of malignant B-cells to migrate to the bone marrow and home into the stromal layer. Therefore, this study aimed to investigate whether stromal cells were also able to inhibit type II anti-CD20 antibody mechanisms of action, contributing to resistance to therapy. Methods: A stromal-tumor co-culture was established in vitro between Raji or Daudi B-cell tumor cells and M210B4 stromal cells in 24 well plates. Results: Contact with stromal cells was able to protect tumor cells from obinutuzumab mediated programmed cell death (PCD), antibody dependent cellular phagocytosis and antibody dependent cellular cytotoxicity. Furthermore, such protection required direct contact between stroma and tumor cells. Stromal cells appeared to interfere with obinutuzumab mediated B-cell homotypic adhesion through inhibiting and reversing actin remodelling, potentially as a result of stromal-tumor cell contact leading to downregulation of CD20 on the surface of tumor cells. Further evidence for the potential role of CD20 downregulation comes through the reduction in surface CD20 expression and inhibition of obinutuzumab mediated PCD when tumor cells are treated with Ibrutinib in the presence of stromal cells. The proteomic analysis of tumor cells after contact with stromal cells led to the identification of a number of altered pathways including those involved in cell adhesion and the actin cytoskeleton and remodeling. Discussion: This work demonstrates that contact between tumor cells and stromal cells leads to inhibition of Obinutuzumab effector functions and has important implications for future therapies to improve outcomes to anti-CD20 antibodies. A deeper understanding of how anti-CD20 antibodies interact with stromal cells could prove a useful tool to define better strategies to target the micro-environment and ultimately improve patient outcomes in B-cell malignancies.

SELECTION OF CITATIONS
SEARCH DETAIL
...