Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (413)

Search Parameters:
Keywords = anti-CD40 monoclonal antibody

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
15 pages, 481 KB  
Review
Bispecific Antibodies: Strategies Available to Optimize Their Safe Delivery in Patients with Multiple Myeloma
by Hannah Victoria Giles and Bhuvan Kishore
Antibodies 2026, 15(1), 5; https://doi.org/10.3390/antib15010005 - 5 Jan 2026
Viewed by 286
Abstract
Bispecific antibodies (BsAbs) have emerged as an important new class drugs for the treatment of multiple myeloma (MM) over the last few years. Currently, BsAbs are only licensed for use as monotherapy in patients with relapsed/refractory MM who have had at least three [...] Read more.
Bispecific antibodies (BsAbs) have emerged as an important new class drugs for the treatment of multiple myeloma (MM) over the last few years. Currently, BsAbs are only licensed for use as monotherapy in patients with relapsed/refractory MM who have had at least three prior lines of treatment and are triple class-exposed (patients who have received an anti-CD38 monoclonal antibody, an immunodulatory drug, and a proteasome inhibitor). However, their use in earlier lines, including in the upfront setting, is being explored in multiple ongoing clinical trials with promising early results. The BsAbs have specific toxicities, including a high rate of low-grade cytokine release syndrome and, less commonly, immune effector cell-associated neurotoxicity syndrome. These immune-related toxicities occur almost exclusively during the initiation phase of the BsAbs. This has led to frequent hospitalization of patients for the duration of the initial step-up dosing phase. Strategies that could facilitate outpatient step-up dosing, such as tocilizumab prophylaxis, will become even more critical if BsAbs move into earlier lines of treatment and are used in larger numbers of patients. Optimizing infection prophylaxis is critical for ensuring the safe delivery of BsAbs as infection is the leading cause of non-relapse mortality in patients being treated with BsAbs. Multiple strategies to minimize the infection risk, including antimicrobial prophylaxis, immunoglobulin replacement, vaccination and reduced dosing frequency, have been evaluated. The clinical data on the efficacy of these supportive measures are described in this review article alongside the available strategies for mitigating and managing CRS and ICANS. Full article
Show Figures

Figure 1

16 pages, 7433 KB  
Article
Two Decades of Real-World Study in Newly Diagnosed Multiple Myeloma: Evolving Treatment and Outcomes in China with Reference to the United States
by Jingyu Xu, Meng Shu, Hsingwen Chung, Jian Cui, Yuntong Liu, Wenqiang Yan, Qirui Bai, Ning Dai, Lingna Li, Jieqiong Zhou, Yating Li, Chenxing Du, Shuhui Deng, Weiwei Sui, Yan Xu, Hong Qiu, Lugui Qiu and Gang An
Cancers 2026, 18(1), 53; https://doi.org/10.3390/cancers18010053 - 24 Dec 2025
Viewed by 458
Abstract
Background: The survival of newly diagnosed multiple myeloma (NDMM) has improved markedly worldwide with the introduction of proteasome inhibitors (PIs), immunomodulatory drugs (IMiDs), and anti-CD38 monoclonal antibodies. However, real-world progress among Chinese patients remains underexplored. This study evaluated 20-year survival trends in [...] Read more.
Background: The survival of newly diagnosed multiple myeloma (NDMM) has improved markedly worldwide with the introduction of proteasome inhibitors (PIs), immunomodulatory drugs (IMiDs), and anti-CD38 monoclonal antibodies. However, real-world progress among Chinese patients remains underexplored. This study evaluated 20-year survival trends in patients with NDMM treated in our institute and benchmarked them against outcomes from the Flatiron Health database in the United States. Patients and methods: Consecutive adults diagnosed with NDMM in our institute between 2003 and 2023 were retrospectively analyzed. U.S. patients were identified from the Flatiron Health database using similar inclusion criteria. Clinical characteristics, first-line regimens, and autologous stem cell transplantation (ASCT) rates were summarized. Overall survival (OS) and progression-free survival (PFS) were estimated by Kaplan–Meier methods. Results: Among 1622 Chinese and 12,582 US patients, median age was 57 vs. 68 years. The median PFS and OS of NDMM patients in our institute was 40.1 months and 99.6 months, respectively. Induction therapy in the NICHE cohort changed markedly from primarily chemo-based therapy to combined PIs + IMIDs-based treatment, whereas these treatments were used much earlier in Flatiron. Uptake of new therapies in China increased rapidly after their inclusion in national health insurance. ASCT utilization was higher overall in China (34.9% vs. 22.1%) but remained lower among patients >65 years (6.7% vs. 12.1%). Conclusions: Two decades of real-world data from a major Chinese myeloma center demonstrate substantial improvements in survival and modernization of NDMM treatment, while highlighting persistent disparities amongst older adults. Full article
(This article belongs to the Section Cancer Causes, Screening and Diagnosis)
Show Figures

Figure 1

18 pages, 2012 KB  
Article
Fab Antibody Fragments to Dog Leukocyte Antigen DR (DLA-DR) Directly Suppress Canine Lymphoma Cell Line Growth In Vitro and in Murine Xenotransplant Model
by Aleksandra Studzińska, Marek Pieczka, Angelika Kruszyńska, Leszek Moniakowski, Anna Urbaniak, Andrzej Rapak and Arkadiusz Miazek
Cancers 2026, 18(1), 48; https://doi.org/10.3390/cancers18010048 - 23 Dec 2025
Viewed by 336
Abstract
Background/Objectives: Canine Diffuse Large B-cell Lymphoma (cDLBCL) is characterized by a high prevalence of MHC II DR (DLA-DR) antigen overexpression. Murine anti-pan-DLA-DR monoclonal antibodies (mAbs) B5 and E11 have been previously observed to promote death of cDLBCL cells in vitro and in vivo. [...] Read more.
Background/Objectives: Canine Diffuse Large B-cell Lymphoma (cDLBCL) is characterized by a high prevalence of MHC II DR (DLA-DR) antigen overexpression. Murine anti-pan-DLA-DR monoclonal antibodies (mAbs) B5 and E11 have been previously observed to promote death of cDLBCL cells in vitro and in vivo. Consequently, DLA-DR antigens are considered a prospective target for passive immunotherapy aside from CD20. While infusion of anti-pan MHC II mAbs has demonstrated tumor suppression in cDLBCL xenografted immunodeficient mice, the relative contributions of direct cellular versus immune-mediated mechanisms to this therapeutic effect remain undefined. This study aimed to dissect these potential mechanisms of mAb E11. Methods: Canine lymphoma and leukemia cell lines CLBL1 and CLB70 were incubated with full E11 antibody or its F(ab′)2 and Fab fragments and cell viability was assessed with sub-G1 assay then, NOD-SCID mice were xenotransplanted with 1.5 × 107 canine CLBL1 cells expressing nanoluciferase and were infused either with mAb E11 or its fragments, each at 1 mg/kg body mass, twice weekly for three consecutive weeks. Tumor burden was monitored by assessing body weight, nanoluciferase activity in blood, and by flow cytometric analyses of bone marrow tumor cell content. Time to tumor progression (TTP) was calculated based on weight loss and luminescence measurements. Results: We observed cytotoxic activity of monovalent E11-Fab fragments in vitro and in vivo. The mean TTP for mice treated with irrelevant mouse IgG antibodies was 9.8 ± 4.65 days. In contrast, treatment with E11 Fab fragments resulted in a TTP of 19.1 ± 2.67 days, which was similar to that achieved with the full E11 mAb (19.5 ± 1.73 days) and E11 F(ab′)2 fragments (18.1 ± 2.9 days). Conclusions: Our findings demonstrate a potent antibody cytotoxicity mechanism that operates in vivo and is independent of cell surface MHC II crosslinking or Fc engagement. These data support the promising potential of E11-Fab fragments for further clinical development as a therapeutic agent in canine lymphoma. Full article
(This article belongs to the Special Issue Advances in B-Cell Lymphoma: From Diagnostics to Cure)
Show Figures

Figure 1

18 pages, 2573 KB  
Systematic Review
Efficacy and Safety of Newly Diagnosed Multiple Myeloma Combination Therapies: A Systematic Review Integrating Network Meta-Analysis and Real-World Vigilance Study
by Yanjun Liu, Ying Zhang, Wenhui Yang, Haoyan Du, Shijie Sun, Zuojing Li and Dongsheng Zong
Pharmaceuticals 2026, 19(1), 18; https://doi.org/10.3390/ph19010018 - 21 Dec 2025
Viewed by 301
Abstract
Background: Although anti-CD38 monoclonal antibody-based regimens are standard care for newly diagnosed multiple myeloma (NDMM), direct comparative efficacy and comprehensive real-world safety data remain scarce. Methods: We conducted a systematic review and Bayesian network meta-analysis (NMA) of randomized controlled trials (RCTs). [...] Read more.
Background: Although anti-CD38 monoclonal antibody-based regimens are standard care for newly diagnosed multiple myeloma (NDMM), direct comparative efficacy and comprehensive real-world safety data remain scarce. Methods: We conducted a systematic review and Bayesian network meta-analysis (NMA) of randomized controlled trials (RCTs). Efficacy was assessed using hazard ratios (HRs) for progression-free survival and odds ratios (ORs) for response rates, with treatment rankings evaluated by Surface Under the Cumulative Ranking (SUCRA) values. Separately, adverse event reports for daratumumab, bortezomib, lenalidomide, and dexamethasone (D_VRd) regimens were extracted from the US FDA Adverse Event Reporting System (FAERS) (Q1 2015–Q2 2025). Statistical analyses were performed using R (4.3.3) and STATA (16.0). Results: The NMA included 33 RCTs. For the primary efficacy endpoints, compared to the standard bortezomib, lenalidomide, and dexamethasone (VRd) regimen, both D_VRd (OR = 3.21, 95% CI: 2.46–4.26; HR = 0.48, 95% CI: 0.38–0.63) and isatuximab plus VRd (Isa_VRd) (OR = 1.71, 95% CI: 1.25–2.32; HR = 0.66, 95% CI: 0.51–0.85) regimens demonstrated superior efficacy. Subsequent pharmacovigilance analysis of D_VRd identified 11,714 FAERS reports, yielding 197 significant adverse drug event signals (64 unlabeled). These signals primarily affected elderly males and showed a bimodal distribution pattern. Conclusions: Combination regimens containing anti-CD38 monoclonal antibodies demonstrate superiority in achieving deep remission and survival benefits, with D_VRd and Isa_VRd regimens showing particularly outstanding performance. However, efficacy and safety profiles vary across different combination regimens. Real-world data analysis further indicates that the D_VRd regimen carries several safety risk signals that remain underappreciated and exhibits a bimodal time distribution pattern. These findings provide new evidence to guide clinical decision-making and risk-stratified monitoring. Full article
(This article belongs to the Section Biopharmaceuticals)
Show Figures

Figure 1

24 pages, 376 KB  
Review
Rituximab in Connective Tissue Disease-Associated Interstitial Lung Disease: From Immunopathogenesis to Therapeutic Implications
by Dimitrios Chatzis, Aggelos Banos, Antonis Fanouriakis, Theodoros Karampitsakos and Vasilios Tzilas
Int. J. Mol. Sci. 2026, 27(1), 46; https://doi.org/10.3390/ijms27010046 - 20 Dec 2025
Viewed by 659
Abstract
Connective tissue disease-associated interstitial lung disease (CTD-ILD) comprises a heterogeneous group of immune-mediated pulmonary disorders with significant morbidity and mortality. The pathogenesis involves complex interactions of autoimmunity, chronic inflammation, and fibrosis. B cells play a central role in these processes through antigen presentation, [...] Read more.
Connective tissue disease-associated interstitial lung disease (CTD-ILD) comprises a heterogeneous group of immune-mediated pulmonary disorders with significant morbidity and mortality. The pathogenesis involves complex interactions of autoimmunity, chronic inflammation, and fibrosis. B cells play a central role in these processes through antigen presentation, autoantibody production, cytokine secretion, and the formation of ectopic lymphoid tissue within the lung parenchyma. Rituximab (RTX)—a chimeric anti-CD20 monoclonal antibody—depletes B cells and has emerged as a promising therapeutic agent for CTD-ILD. This review comprehensively presents the immunopathogenic mechanisms underlying CTD-ILD, elaborating on the multifaceted mode of action of RTX and summarizing the evolving clinical evidence. Full article
22 pages, 4641 KB  
Article
Radionuclide-Dependent Stimulation of Antitumor Immunity in GD2-Targeted Radiopharmaceutical Therapy Combined with Immune Checkpoint Inhibitors
by Cynthia Lilieholm, Jen Zaborek, Ohyun Kwon, Adedamola O. Adeniyi, Caroline P. Kerr, Hansel Comas Rojas, Malick Bio Idrissou, Carolina A. Ferreira, Paul A. Clark, Won Jong Jin, Joseph J. Grudzinski, Amy K. Erbe, Eduardo Aluicio-Sarduy, Thines Kanagasundaram, Justin J. Wilson, Jonathan W. Engle, Reinier Hernandez, Bryan Bednarz, Zachary S. Morris and Jamey P. Weichert
Radiation 2025, 5(4), 39; https://doi.org/10.3390/radiation5040039 - 9 Dec 2025
Viewed by 630
Abstract
Radiopharmaceutical therapy (RPT) offers tumor-selective radiation delivery and represents a promising platform for combination with immune checkpoint inhibitors (ICIs). While prior studies suggest that RPT can stimulate antitumor immunity, synergy with ICIs may depend on radionuclide properties, absorbed dose, and radiation distribution within [...] Read more.
Radiopharmaceutical therapy (RPT) offers tumor-selective radiation delivery and represents a promising platform for combination with immune checkpoint inhibitors (ICIs). While prior studies suggest that RPT can stimulate antitumor immunity, synergy with ICIs may depend on radionuclide properties, absorbed dose, and radiation distribution within the tumor microenvironment. This study evaluated how radionuclide selection and dose influence immune stimulation and therapeutic efficacy of GD2-targeted antibody-based RPT combined with ICIs. Dinutuximab, an anti-GD2 monoclonal antibody, was radiolabeled with β-emitters (90Y, 177Lu) or an α-emitter (225Ac). C57Bl6 mice bearing GD2+ tumors received 4 or 15 Gy tumor-absorbed doses, determined by individualized dosimetry, with or without dual ICIs (anti-CTLA-4 and anti-PD-L1). In vivo imaging, ex vivo biodistribution, survival, histological, and gene expression analyses were performed to assess therapeutic and immunological outcomes. All radiolabeled constructs demonstrated preferential uptake in GD2+ tumors. Combination therapy improved survival in a radionuclide- and dose-dependent manner, with the greatest benefit in the 225Ac + ICI group at 15 Gy. Treatment activated type I interferon signaling and increased MHC-I and PD-L1 expression. Notably, 90Y reduced regulatory T cells, enhancing CD8+/Treg ratios, while 225Ac induced robust interferon-driven activation. Radionuclide selection and absorbed dose critically shape immune and therapeutic outcomes of antibody-based RPT combined with ICIs, underscoring the importance of delivery mechanism and dose optimization in combination therapy strategies. Full article
Show Figures

Figure 1

14 pages, 652 KB  
Review
The Inflammatory Role of Serum Amyloid A in the Pathogenesis and Progression of Diabetic Nephropathy
by Antigoni Stavrou, Christina A. Kousparou and Argyrios Tsakalis
J. Clin. Med. 2025, 14(23), 8427; https://doi.org/10.3390/jcm14238427 - 27 Nov 2025
Viewed by 1108
Abstract
Diabetic nephropathy (DN) remains the leading cause of end-stage renal disease (ESRD) worldwide, primarily affecting individuals with Type 2 Diabetes Mellitus (T2DM). While traditional risk factors—such as hypertension, poor glycemic control, and dyslipidemia—are well known, recent research has illuminated the pivotal role of [...] Read more.
Diabetic nephropathy (DN) remains the leading cause of end-stage renal disease (ESRD) worldwide, primarily affecting individuals with Type 2 Diabetes Mellitus (T2DM). While traditional risk factors—such as hypertension, poor glycemic control, and dyslipidemia—are well known, recent research has illuminated the pivotal role of inflammation in DN pathogenesis. Inflammatory processes involving chemokines, cytokines, immune cell infiltration, and pro-fibrotic signaling pathways (e.g., NFκB, JAK/STAT) contribute significantly to glomerular and tubulointerstitial damage. Key immune players include macrophages and T lymphocytes, particularly CD4+ T cells, which correlate with disease severity and progression. Serum Amyloid A (SAA), an acute-phase reactant traditionally associated with Serum Amyloid A Amyloidosis (AA amyloidosis), has emerged as both a biomarker and active mediator of renal inflammation in DN. SAA promotes cytokine release, leukocyte recruitment, and extracellular matrix remodeling, contributing to glomerular and tubular injury. Elevated Saa3 expression in experimental models correlates with DN progression, while activation of the advanced glycation end products and the receptors for advanced glycation end products (AGE–RAGE) axis in podocytes enhances SAA upregulation and inflammatory signaling. Increasing evidence now indicates that SAA functions, not only as a marker of systemic inflammation, but also as a mechanistically significant driver of intrarenal injury, bridging metabolic dysregulation with sustained inflammatory and fibrotic signaling. Emerging therapeutic approaches—including interleukin 6 (IL-6) blockade, inhibition of AGE formation, targeted anti-fibrotic agents, and recently developed SAA-directed RNA or peptide therapeutics—underscore the therapeutic potential of modulating SAA activity in DN. Preclinical evidence further supports the efficacy of monoclonal antibodies, signaling inhibitors, and dietary anti-inflammatory compounds in mitigating renal injury. Collectively, these developments position SAA as a central mediator at the intersection of metabolic, inflammatory, and fibrotic pathways, highlighting its promise as both a diagnostic biomarker and a therapeutic target for early intervention in diabetic kidney disease. Full article
(This article belongs to the Section Nephrology & Urology)
Show Figures

Figure 1

20 pages, 1223 KB  
Review
Mechanistic Insights and Advances of Bispecific T Cell Engaging Antibodies Therapy in Multiple Myeloma
by Ting Fang Tang, Chin Sum Cheong, Chung Yeng Looi, Won Fen Wong and Gin Gin Gan
Medicina 2025, 61(12), 2113; https://doi.org/10.3390/medicina61122113 - 27 Nov 2025
Viewed by 1560
Abstract
Multiple myeloma (MM) is a clonal malignancy of terminally differentiated plasma cells characterized by bone marrow infiltration and excessive production of monoclonal immunoglobulins, leading to end-organ damage such as osteolytic bone lesions. Despite substantial therapeutic progress achieved with proteasome inhibitors, immunomodulatory drugs, and [...] Read more.
Multiple myeloma (MM) is a clonal malignancy of terminally differentiated plasma cells characterized by bone marrow infiltration and excessive production of monoclonal immunoglobulins, leading to end-organ damage such as osteolytic bone lesions. Despite substantial therapeutic progress achieved with proteasome inhibitors, immunomodulatory drugs, and anti-CD38 monoclonal antibodies, multiple myeloma remains incurable, and outcomes for triple-class-refractory patients remain dismal, with median survival below one year. Bispecific T cell engaging antibodies (TCEs) have recently emerged as a promising immunotherapeutic approach capable of redirecting cytotoxic T cells to eliminate malignant plasma cells. These engineered antibodies simultaneously engage CD3 on T cells and a tumor-associated antigen such as B cell maturation antigen (BCMA), G protein-coupled receptor family C group 5 member D (GPRC5D), or Fc receptor homolog 5 (FcRH5), thereby forming an immune synapse that triggers T cell activation, cytokine secretion, and perforin–granzyme-mediated apoptosis of the targeted B cell. This review summarizes the molecular design, mechanism of action, and clinical development of TCEs in MM, encompassing early bi-specific T cell engagers (BiTE) constructs such as AMG 420 and next-generation IgG-like molecules including teclistamab. Pivotal clinical trials have demonstrated overall response rates between 43% and 73%, accompanied by durable remissions and manageable safety profiles. Future directions include earlier-line integration, synergistic combinations with immunomodulatory or costimulatory agents, and the development of trispecific formats to overcome antigen escape and T cell exhaustion. Collectively, TCEs represent a paradigm shift toward durable, immune-mediated disease control in multiple myeloma. Full article
Show Figures

Figure 1

14 pages, 2315 KB  
Article
Therapeutic Potential of an Anti-CD44v6 Monoclonal Antibody in Xenograft Models of Colorectal and Gastric Cancer
by Aoi Hirayama, Tomohiro Tanaka, Tomokazu Ohishi, Keisuke Shinoda, Takuya Nakamura, Airi Nomura, Naoki Kojo, Haruto Araki, Kaito Suzuki, Mika K. Kaneko, Hiroyuki Suzuki and Yukinari Kato
Cells 2025, 14(23), 1873; https://doi.org/10.3390/cells14231873 - 26 Nov 2025
Viewed by 668
Abstract
CD44 variant (CD44v) isoforms are involved in promoting cancer metastasis, sustaining cancer stem cell (CSC) properties, and conferring resistance to therapeutic interventions. Consequently, the development of monoclonal antibodies (mAbs) targeting CD44v represents a crucial strategy for eliminating CD44v-positive cancer cells. Previously, an anti-CD44v6 [...] Read more.
CD44 variant (CD44v) isoforms are involved in promoting cancer metastasis, sustaining cancer stem cell (CSC) properties, and conferring resistance to therapeutic interventions. Consequently, the development of monoclonal antibodies (mAbs) targeting CD44v represents a crucial strategy for eliminating CD44v-positive cancer cells. Previously, an anti-CD44v6 mAb, C44Mab-9 (mouse IgG1, κ), was established. C44Mab-9 recognizes explicitly the epitope encoded by the variant exon 6-encoded region of CD44 and applies to flow cytometry, western blotting, and immunohistochemistry. To assess the therapeutic potential, a mouse IgG2a isotype of C44Mab-9 (designated C44Mab-9-mG2a) was generated, and the in vitro and in vivo antitumor activities were evaluated using gastric and colorectal cancer cell lines. C44Mab-9-mG2a demonstrated specific binding to CD44v3–10-overexpressed Chinese hamster ovary cells (CHO/CD44v3–10), as well as gastric cancer (NUGC-4) and colorectal cancer (COLO201 and COLO205) in flow cytometry. C44Mab-9-mG2a exerted antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) against CHO/CD44v3–10, NUGC-4, COLO201, and COLO205. Moreover, systemic administration of C44Mab-9-mG2a significantly inhibited tumor growth in CHO/CD44v3–10, NUGC-4, COLO201, and COLO205 xenografts compared with the control IgG2a. These findings indicate that C44Mab-9-mG2a could be applied to the mAb-based therapy against CD44v6-positive tumors. Full article
(This article belongs to the Special Issue Cellular Mechanisms of Anti-Cancer Therapies)
Show Figures

Figure 1

38 pages, 1175 KB  
Review
Novel Therapeutic Approaches in Pediatric Acute Lymphoblastic Leukemia
by Maria Maddalena Marrapodi, Alessandra Di Paola, Giuseppe Di Feo, Oriana Di Domenico, Martina Di Martino, Lucia Argenziano, Marianna Falcone, Daniela Di Pinto, Francesca Rossi and Elvira Pota
Int. J. Mol. Sci. 2025, 26(23), 11362; https://doi.org/10.3390/ijms262311362 - 24 Nov 2025
Viewed by 1218
Abstract
Acute lymphoblastic leukemia (ALL) is the most common pediatric malignancy, characterized by the clonal proliferation of immature lymphoid precursors. The distinction between B-cell ALL (B-ALL) and T-cell ALL (T-ALL) is fundamental, as each subtype exhibits distinct cytomorphological, genetic, and clinical features influencing prognosis [...] Read more.
Acute lymphoblastic leukemia (ALL) is the most common pediatric malignancy, characterized by the clonal proliferation of immature lymphoid precursors. The distinction between B-cell ALL (B-ALL) and T-cell ALL (T-ALL) is fundamental, as each subtype exhibits distinct cytomorphological, genetic, and clinical features influencing prognosis and therapeutic strategies. Conventional multi-phase chemotherapy has significantly improved survival rates, yet its efficacy is limited by severe short- and long-term toxicities, highlighting the need for more selective therapeutic approaches. Advances in molecular profiling have enabled the identification of key oncogenic pathways, paving the way for targeted therapies such as tyrosine kinase inhibitors (TKIs), JAK-STAT pathway inhibitors, BCL-2 antagonists, and agents modulating epigenetic and cell cycle regulators. Concurrently, immunotherapeutic strategies have transformed the therapeutic landscape of pediatric ALL. Bispecific antibodies such as blinatumomab (anti-CD19), antibody–drug conjugates like inotuzumab ozogamicin (anti-CD22), and monoclonal antibodies such as daratumumab (anti-CD38) have demonstrated efficacy in relapsed or refractory disease with improved safety profiles. Moreover, CAR-T-cell therapy, particularly CD19-directed products, has shown unprecedented remission rates in refractory B-ALL. The integration of targeted and immune-based therapies into conventional regimens represents a decisive step toward precision medicine, aiming to enhance survival outcomes while reducing treatment-related toxicity and improving quality of life in ALL children. This review aims to provide a comprehensive overview of the current understanding of ALL pathobiology and therapeutic approaches, with particular emphasis on the expanding role of immunotherapeutic strategies in pediatric disease. Full article
(This article belongs to the Special Issue Molecular Advances in Pediatric Diseases)
Show Figures

Graphical abstract

14 pages, 675 KB  
Article
Assessing Peripheral Blood Biomarkers and Predictive Patterns in Multiple Sclerosis Using Cytokines and Immune Gene Expression Profiles in Ocrelizumab-Treated Patients: Tracking Tumor Necrosis Factor
by Bojan Jevtić, Nikola Momcilovic, Goran Stegnjaić, Milica Lazarević, Suzana Stanisavljević, Olivera Tamas, Nikola Veselinovic, Maja Budimkic, Sarlota Mesaros, Đorđe Miljković, Tatjana Pekmezovic, Jelena Drulovic and Neda Nikolovski
Int. J. Mol. Sci. 2025, 26(23), 11295; https://doi.org/10.3390/ijms262311295 - 22 Nov 2025
Viewed by 581
Abstract
Ocrelizumab, a humanized monoclonal anti-CD20 and B cell-depleting antibody, is a disease-modifying therapy for multiple sclerosis (MS), a chronic inflammatory, demyelinating, and neurodegenerative disease of the central nervous system. However, reliable predictive biomarkers of ocrelizumab’s effectiveness, such as cytokine expression profiles in peripheral [...] Read more.
Ocrelizumab, a humanized monoclonal anti-CD20 and B cell-depleting antibody, is a disease-modifying therapy for multiple sclerosis (MS), a chronic inflammatory, demyelinating, and neurodegenerative disease of the central nervous system. However, reliable predictive biomarkers of ocrelizumab’s effectiveness, such as cytokine expression profiles in peripheral blood mononuclear cells (PBMCs), are lacking. The aim of this study was to identify immunological biomarkers of ocrelizumab treatment response in MS patients, during a two-year follow-up. mRNA expression for specific immune molecules in PBMCs was measured, and consequently correlated with the clinical and radiological parameters of disease activity. PBMCs were obtained from 80 MS patients (35 with relapsing–remitting MS-RRMS and 45 with primary progressive-PPMS), immediately before initiating ocrelizumab treatment and thereafter every 6 months (before the administration of the next dose of ocrelizumab). Expression of the B cell marker CD19; the pro-inflammatory cytokines interleukin (IL)1B, IL6, and tumor necrosis factor (TNF); and a costimulatory cell marker CD86 were determined. In both RMS and PPMS patients treated with ocrelizumab, higher baseline expression of TNF was statistically significantly associated with an increased risk of developing evidence of disease activity and a greater likelihood of disability progression, at month 24. This result implies that PBMCs’ TNF mRNA expression might be potentially considered as a prognostic biomarker of ocrelizumab effectiveness in MS patients. However, further studies comprising large cohorts and additional immunological parameters are warranted. Full article
Show Figures

Graphical abstract

14 pages, 637 KB  
Article
The Strange Case of Functional High-Risk Multiple Myeloma Patients: Is It Possible to Identify Them in Clinical Practice?
by Sonia Morè, Massimo Offidani, Laura Corvatta, Tommaso Za, Francesca Fazio, Martina Gherardini, Velia Bongarzoni, Barbara Anaclerico, Luca Franceschini, Silvia Ferraro, Luca Cupelli, Carmine Liberatore, Laura De Padua, Angela Rago, Silvia Gentili, Roberto Latagliata, Mariagrazia Garzia, Iole Cordone, Valeria Mezzanotte, Elena Rossi, Francesca Di Landro, Maria Zaira Limongi, Erika Morsia, Antonella Poloni and Maria Teresa Petrucciadd Show full author list remove Hide full author list
Cancers 2025, 17(21), 3580; https://doi.org/10.3390/cancers17213580 - 6 Nov 2025
Viewed by 749
Abstract
Background: Early relapse in multiple myeloma (MM) is a major predictor of poor prognosis, regardless of cytogenetic risk or treatment intensity. Methods: Here we analyzed 1026 MM patients treated across 12 Italian hematology centers. FHR was defined as progression-free survival (PFS) ≤18 months [...] Read more.
Background: Early relapse in multiple myeloma (MM) is a major predictor of poor prognosis, regardless of cytogenetic risk or treatment intensity. Methods: Here we analyzed 1026 MM patients treated across 12 Italian hematology centers. FHR was defined as progression-free survival (PFS) ≤18 months in transplant-eligible (TE) and ≤12 months in non-transplant-eligible (NTE) patients. Logistic regression and ROC analysis were used to identify significant predictors of FHR and build a risk score. Results: FHR status was identified in 175 patients (17%). These patients had significantly shorter PFS (7 vs. 57.5 months) and overall survival (19 months vs. not reached; p < 0.001). FHR status was associated with higher median LDH, lower Hb level, higher creatinine level and lower platelets count. Modified EASIX formula was built by these significant continuous variables, to be tested in a logistic analysis: [(LDH × creatinine)/(Hb × PLT) × 100]. A significantly higher rate of FHR was found with a score > 2.0 (89% vs. 11%, p < 0.001). Multivariate logistic analysis selected the above formula, ECOG PS ≥ 2 and ISS III as factors associated with FHR. Scoring these variables according to OR, three groups of patients were segregated with a rate of FHR patients of 7%, 29.5%, and 63.5%, respectively. Treatment with anti-CD38 monoclonal antibodies was associated with lower FHR frequency. Conclusions: This study proposes a simple, clinically applicable model to identify FHR MM patients early in their disease course. However, very in-depth biological tools, not available in clinical practice, are needed to identify singularly risk of becoming FHR. Full article
(This article belongs to the Section Clinical Research of Cancer)
Show Figures

Figure 1

13 pages, 1088 KB  
Article
Inflammatory Biomarkers for Thrombotic Risk Assessment in Multiple Myeloma Patients on IMiD/aCD38-Based Regimens: Insights from a Prospective Observational Study
by Cirino Botta, Anna Maria Corsale, Claudia Cammarata, Fabiana Di Fazio, Emilia Gigliotta, Andrea Rizzuto, Manuela Ingrascì, Maria Speciale, Cristina Aquilina, Marta Biondo, Andrea Romano, Mariasanta Napolitano, Marta Mattana and Sergio Siragusa
Biomolecules 2025, 15(11), 1533; https://doi.org/10.3390/biom15111533 - 31 Oct 2025
Viewed by 749
Abstract
Thrombosis is a common complication in multiple myeloma (MM) patients treated with immunomodulatory drugs (IMiDs), including thalidomide, lenalidomide, and pomalidomide. When combined with anti-CD38 monoclonal antibodies, these agents are highly effective but may increase thrombotic events (TE), potentially delaying therapy. This exploratory, hypothesis-generating [...] Read more.
Thrombosis is a common complication in multiple myeloma (MM) patients treated with immunomodulatory drugs (IMiDs), including thalidomide, lenalidomide, and pomalidomide. When combined with anti-CD38 monoclonal antibodies, these agents are highly effective but may increase thrombotic events (TE), potentially delaying therapy. This exploratory, hypothesis-generating analysis, conducted within the MMVision mono-institutional prospective study, included 53 MM patients who initiated IMiD plus anti-CD38 therapy between May 2021 and December 2022 (median follow-up: 18 months). Treatment regimens comprised lenalidomide (n = 36) or thalidomide (n = 15) with daratumumab, and pomalidomide (n = 2) with isatuximab. Most patients (n = 38) received frontline therapy, and all were given thromboprophylaxis according to guidelines, mainly aspirin (73%). Five patients (9.4%) developed VTE after a median of 48 days, managed with short-term low-molecular-weight heparin (LMWH). Exploratory analysis of 27 clinical/laboratory parameters suggested possible associations between VTE and low levels of beta-2 microglobulin, ferritin, intact/free lambda light chains, and monocyte-to-lymphocyte ratio. Notably, four of the five VTEs occurred in patients without lytic bone disease, typically associated with bone-driven inflammation in MM. Although all patients received aspirin prophylaxis from treatment initiation, it remains unclear whether thrombosis would also have occurred among those with higher inflammatory burden. These preliminary observations may indicate that in patients with relatively lower inflammation, aspirin prophylaxis could be less effective, potentially favoring VTE onset. In two VTE cases, cytokine profiling showed decreased M-CSF, SCLF-β, and MIP-1α, with increased G-CSF, raising the hypothesis of distinct immune-inflammatory pathways contributing to TEs. Given the limited number of patients and thrombotic events, and the cytokine data available for only two VTE cases, these associations should be regarded as exploratory and interpreted with caution. Overall, these exploratory findings warrant validation in larger, independent cohorts and may help generate hypotheses on how inflammatory signatures influence thrombotic risk and prophylaxis efficacy in MM patients receiving IMiD/anti-CD38-based regimens. Full article
Show Figures

Figure 1

15 pages, 1014 KB  
Systematic Review
Efficacy and Safety of Isatuximab Combination Therapy in Multiple Myeloma: A Meta-Analysis of Randomized Controlled Trials
by Chi Wang, Zhengyang Xu, Meilin Jiang, Yuzhe Chen and Yu Lan
Cancers 2025, 17(21), 3494; https://doi.org/10.3390/cancers17213494 - 30 Oct 2025
Viewed by 1060
Abstract
Background: This meta-analysis evaluates the efficacy and safety of isatuximab, an anti-CD38 monoclonal antibody, in combination regimens for newly diagnosed (NDMM) and relapsed/refractory multiple myeloma (RRMM). Methods: We systematically searched major databases for randomized controlled trials (RCTs) comparing isatuximab-based therapy with standard regimens [...] Read more.
Background: This meta-analysis evaluates the efficacy and safety of isatuximab, an anti-CD38 monoclonal antibody, in combination regimens for newly diagnosed (NDMM) and relapsed/refractory multiple myeloma (RRMM). Methods: We systematically searched major databases for randomized controlled trials (RCTs) comparing isatuximab-based therapy with standard regimens up to September 2025. Efficacy and safety analyses were performed separately for NDMM and RRMM populations using random-effects models. Efficacy outcomes included progression-free survival (PFS), overall survival (OS), overall response rate (ORR), very good partial response (VGPR) or better, and minimal residual disease (MRD) negativity rate. Safety was assessed by grade ≥ 3 adverse events. Results: In NDMM patients, isatuximab significantly improved PFS (HR = 0.66, 95% CI: 0.52–0.84, p = 0.001) and MRD negativity rates (RR = 1.28, 95% CI: 1.13–1.45, p < 0.001), but not OS (HR = 1.01, p = 0.937), ORR (RR = 1.02, p = 0.49), or VGPR or better (RR = 1.10, p = 0.13). In RRMM patients, isatuximab significantly improved PFS (HR = 0.61, 95% CI: 0.50–0.74, p < 0.001) and showed strong trends favoring OS (HR = 0.81, 95% CI: 0.65–1.00, p = 0.051) and ORR (RR = 1.30, 95% CI: 0.79–2.16, p = 0.303), while significantly increasing MRD negativity (RR = 4.37, 95% CI: 0.60–31.68, p = 0.144). A trend toward improved OS was observed in RRMM (HR = 0.81, p = 0.051). In NDMM, PFS benefit was significant for standard-risk but not high-risk cytogenetics. Safety analysis showed an increased risk of grade ≥ 3 adverse events RRMM (RR = 1.18, p < 0.001) but not in NDMM (RR = 1.08, p = 0.064), primarily driven by neutropenia (NDMM RR = 1.96, p = 0.003; RRMM RR = 1.77, p = 0.039) and pneumonia in NDMM (RR = 1.80, p = 0.001). Conclusion: Isatuximab-based regimens significantly improve PFS and depth of response with a manageable safety profile, supporting its use across MM settings, though efficacy in NDMM may vary by cytogenetic risk. Full article
(This article belongs to the Special Issue Multiple Myeloma: Diagnosis and Therapy)
Show Figures

Figure 1

14 pages, 1056 KB  
Article
Cytokine Dynamics During Ustekinumab Induction as Predictors of Treatment Response in Crohn’s Disease: An Observational Study
by Alejandro Mínguez, Beatriz Mateos, Marisa Iborra, Mariam Aguas, Guillermo Bastida, Alejandro Garrido, Elena Cerrillo, Sonia García, Lluís Tortosa, Inés Moret and Pilar Nos
Biomedicines 2025, 13(11), 2608; https://doi.org/10.3390/biomedicines13112608 - 24 Oct 2025
Viewed by 825
Abstract
Background/Objectives: Crohn’s disease (CD) is a chronic immune-mediated disorder with heterogeneous response to biologic therapies. Ustekinumab (UST), an anti-IL-12/23 monoclonal antibody, is effective in CD, but predictive biomarkers of treatment response remain lacking. This study aimed to investigate cytokine dynamics during UST [...] Read more.
Background/Objectives: Crohn’s disease (CD) is a chronic immune-mediated disorder with heterogeneous response to biologic therapies. Ustekinumab (UST), an anti-IL-12/23 monoclonal antibody, is effective in CD, but predictive biomarkers of treatment response remain lacking. This study aimed to investigate cytokine dynamics during UST induction and to evaluate their association with clinical and biochemical outcomes in an observational cohort of CD patients. Methods: We prospectively recruited 31 adult patients with moderate-to-severe active CD initiating UST therapy at a tertiary referral center. Peripheral blood and stool samples were collected at baseline and weeks 4, 8, and 16. UST trough concentrations, C-reactive protein (CRP), fecal calprotectin (FC), hemoglobin, albumin, and 13 serum cytokines (including IL-1β, IL-6, IL-8, IL-10, IL-12p70, IL-13, IL-17, IL-23, TNF-α, and OSM) were analyzed. Response was defined as a ≥70% reduction in FC at week 16, or, alternatively, CRP < 5 mg/L or a Harvey–Bradshaw Index < 3. Results: Eighteen patients (58%) achieved response at week 16. Responders showed significant reductions in FC, CRP, and disease activity, while non-responders exhibited limited biochemical improvement. Overall, UST induction was associated with a global decrease in proinflammatory cytokines, particularly TNF-α and IL-1β. Responders displayed distinct cytokine patterns, with higher IL-13 levels at week 8 and lower IL-8 concentrations at week 16 compared with non-responders. UST trough levels tended to be higher in responders, and inverse correlations were observed between drug concentrations and several cytokines, including IL-6, IL-8, IL-13, and IL-23. Conclusions: UST induction leads to measurable immunological changes in CD, with differential cytokine dynamics distinguishing responders from non-responders. These findings support the potential of cytokine signatures, in combination with therapeutic drug monitoring, as pharmacodynamic biomarkers to optimize personalized treatment strategies in CD. Full article
Show Figures

Graphical abstract

Back to TopTop