[go: up one dir, main page]

 
 
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

Journals

Article Types

Countries / Regions

Search Results (39)

Search Parameters:
Keywords = alemtuzumab

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 368 KiB  
Review
Current and Emerging Treatment Options in Pediatric Onset Multiple Sclerosis
by Artemis Mavridi, Maria Eleni Bompou, Aine Redmond, Paraschos Archontakis-Barakakis, George D. Vavougios, Dimos D. Mitsikostas and Theodoros Mavridis
Sclerosis 2024, 2(2), 88-107; https://doi.org/10.3390/sclerosis2020007 - 1 Apr 2024
Viewed by 2786
Abstract
Pediatric onset multiple sclerosis (POMS), characterized by the onset of multiple sclerosis before the age of 18, is gaining increased recognition. Approximately 5 percent of MS cases manifest before the age of 18, with less than 1 percent occurring before the age of [...] Read more.
Pediatric onset multiple sclerosis (POMS), characterized by the onset of multiple sclerosis before the age of 18, is gaining increased recognition. Approximately 5 percent of MS cases manifest before the age of 18, with less than 1 percent occurring before the age of 10. Despite its rarity, pediatric MS exhibits distinct characteristics, with an association between younger age at onset and a comparatively slower disease progression. Despite this slower progression, individuals with POMS historically reach disability milestones at earlier ages than those with adult-onset multiple sclerosis. While various immunomodulatory agents demonstrate significant benefits in MS treatment, such as reduced relapse rates and slower accumulation of brain lesions on magnetic resonance imaging (MRI), the majority of disease-modifying therapies (DMTs) commonly used in adult MS lack evaluation through pediatric clinical trials. Current evidence is predominantly derived from observational studies. This comprehensive review aims to consolidate existing knowledge on the mechanisms of action, efficacy, safety profiles, and recommended dosages of available DMTs specifically in the context of pediatric MS. Furthermore, this review outlines recent advancements and explores potential medications still in developmental stages, providing a thorough overview of the current landscape and future prospects for treating POMS. Full article
15 pages, 606 KiB  
Review
Headache in Multiple Sclerosis: A Narrative Review
by Bożena Adamczyk, Natalia Morawiec, Sylwia Boczek, Karol Dańda, Mikołaj Herba, Aleksandra Spyra, Agata Sowa, Jarosław Szczygieł and Monika Adamczyk-Sowa
Medicina 2024, 60(4), 572; https://doi.org/10.3390/medicina60040572 - 30 Mar 2024
Viewed by 1645
Abstract
Background: Multiple sclerosis (MS) is a chronic inflammatory demyelinating disorder of the central nervous system characterized by autoimmune-mediated damage to oligodendrocytes and subsequent myelin destruction. Clinical implications: Clinically, the disease presents with many symptoms, often evolving over time. The insidious onset [...] Read more.
Background: Multiple sclerosis (MS) is a chronic inflammatory demyelinating disorder of the central nervous system characterized by autoimmune-mediated damage to oligodendrocytes and subsequent myelin destruction. Clinical implications: Clinically, the disease presents with many symptoms, often evolving over time. The insidious onset of MS often manifests with non-specific symptoms (prodromal phase), which may precede a clinical diagnosis by several years. Among them, headache is a prominent early indicator, affecting a significant number of MS patients (50–60%). Results: Headache manifests as migraine or tension-type headache with a clear female predilection (female-male ratio 2-3:1). Additionally, some disease-modifying therapies in MS can also induce headache. For instance, teriflunomide, interferons, ponesimod, alemtuzumab and cladribine are associated with an increased incidence of headache. Conclusions: The present review analyzed the literature data on the relationship between headache and MS to provide clinicians with valuable insights for optimized patient management and the therapeutic decision-making process. Full article
(This article belongs to the Section Neurology)
Show Figures

Figure 1

Figure 1
<p>The similarities between MS and migraine headache pathogeneses.</p>
Full article ">
16 pages, 1589 KiB  
Review
Histopathological Markers for Target Therapies in Primary Cutaneous Lymphomas
by Benedetta Sonego, Adalberto Ibatici, Giulia Rivoli, Emanuele Angelucci, Simona Sola and Cesare Massone
Cells 2023, 12(22), 2656; https://doi.org/10.3390/cells12222656 - 20 Nov 2023
Cited by 1 | Viewed by 1479
Abstract
In recent years, targeted (biological) therapies have become available also for primary cutaneous T-cell lymphomas (PCTCLs) including anti-CD30 (brentuximab vedotin) in mycosis fungoides, primary cutaneous anaplastic large T-cell lymphoma, lymphomatoid papulosis; anti-CCR4 (mogamulizumab) in Sezary syndrome; anti-CD123 (tagraxofusp) in blastic plasmocytoid cell neoplasm. [...] Read more.
In recent years, targeted (biological) therapies have become available also for primary cutaneous T-cell lymphomas (PCTCLs) including anti-CD30 (brentuximab vedotin) in mycosis fungoides, primary cutaneous anaplastic large T-cell lymphoma, lymphomatoid papulosis; anti-CCR4 (mogamulizumab) in Sezary syndrome; anti-CD123 (tagraxofusp) in blastic plasmocytoid cell neoplasm. Moreover, anti-PD1 (nivolumab), anti-PDL1 (pembrolizumab, atezolizumab), anti-CD52 (alemtuzumab), anti-KIR3DL2-CD158k (lacutamab), and anti-CD70 (cusatuzumab) have been tested or are under investigations in phase II trials. The expression of these epitopes on neoplastic cells in skin biopsies or blood samples plays a central role in the management of PCTCL patients. This narrative review aims to provide readers with an update on the latest advances in the newest therapeutic options for PCTCLs. Full article
(This article belongs to the Section Cell Signaling)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>(<b>A</b>) MF with LCT: multiple plaques and nodules, partly ulcerated on the trunk. (<b>B</b>) Large neoplastic lymphocytes, partly immunoblastic in appearance with round to oval nuclei and prominent central nucleoli. (<b>C</b>) Strong CD30 positivity (40×). (<b>D</b>) Strong CD4 positivity (10×). (<b>E</b>) High Ki67 expression (100×).</p>
Full article ">Figure 2
<p>(<b>A</b>) SS: diffuse erythema on the trunk. (<b>B</b>) Atypical epidermotropic lymphocytes mainly collected in the typical Pautrier’s (Darier’s) microabscess; band-like infiltrate in the superficial dermis (H&amp;E; 40×). (<b>C</b>) Strong CD30 positivity (40×).</p>
Full article ">
12 pages, 6936 KiB  
Case Report
Long-Smoldering T-prolymphocytic Leukemia: A Case Report and a Review of the Literature
by Hilde K. Gjelberg, Lars Helgeland, Knut Liseth, Francesca Micci, Miriam Sandnes, Hege G. Russnes and Håkon Reikvam
Curr. Oncol. 2023, 30(11), 10007-10018; https://doi.org/10.3390/curroncol30110727 - 18 Nov 2023
Viewed by 2131
Abstract
T-prolymphocytic leukemia (T-PLL) is a rare malignancy of mature T-cells with distinct clinical, cytomorphological, and molecular genetic features. The disease typically presents at an advanced stage, with marked leukocytosis, B symptoms, hepatosplenomegaly, and bone marrow failure. It usually follows an aggressive course from [...] Read more.
T-prolymphocytic leukemia (T-PLL) is a rare malignancy of mature T-cells with distinct clinical, cytomorphological, and molecular genetic features. The disease typically presents at an advanced stage, with marked leukocytosis, B symptoms, hepatosplenomegaly, and bone marrow failure. It usually follows an aggressive course from presentation, and the prognosis is often considered dismal; the median overall survival is less than one year with conventional chemotherapy. This case report describes a patient with T-PLL who, after an unusually protracted inactive phase, ultimately progressed to a highly invasive, organ-involving disease. After initial treatments failed, a novel treatment approach resulted in a significant response. Full article
(This article belongs to the Special Issue Haematological Neoplasms: Diagnosis and Management)
Show Figures

Figure 1

Figure 1
<p>Peripheral blood smear at referral (<b>a</b>) and after 6.5 years of observation (<b>b</b>), with small- to medium-sized lymphoid cells having round nuclei, some with nucleoli (white arrows). Some of the lymphoid cells demonstrate small cytoplasmatic protrusions (“blebs”; black arrows).</p>
Full article ">Figure 2
<p>Photomicrograph of the bone marrow at referral: (<b>a</b>) H&amp;E stain and (<b>b</b>) immunohistochemical staining for T-cell marker CD3 (32×).</p>
Full article ">Figure 3
<p>Timeline illustrating the leukocyte count, hemoglobin level, platelet count, and lactate dehydrogenase level corresponding to clinical events.</p>
Full article ">Figure 4
<p>Flow cytometry immunophenotyping of peripheral blood: (<b>a</b>) The neoplastic T-cells demonstrate bright CD3 and CD5 expression (red dots); reactive T-cells demonstrate dimmer CD5 expression (blue dots). (<b>b</b>) The neoplastic T-cells are predominantly CD4-positive, a subset is CD8-positive, and they are strongly CD52-positive (<b>c</b>). (<b>d</b>) The neoplastic T-cells are TCL1-negative. (Brown dots are granulocytes, and yellow dots are monocytes. The larger dots indicate the median value of the population with the identical color).</p>
Full article ">Figure 5
<p>Photomicrograph of the bone marrow after 6.5 years of observation: (<b>a</b>) H&amp;E (32×). (<b>b</b>) Reticulin stain (25×). (<b>c</b>,<b>d</b>) Immunohistochemical stains for CD3 and CD4 (32×). (<b>e</b>,<b>f</b>) Immunohistochemical stains for TCL1 and CD20 (32×).</p>
Full article ">Figure 6
<p>Cytogenetic analysis of the tumor cells demonstrating a complex karyotype: 47,XY,del(2)(p21),del(5)(q11q13),+8,del(11)(q23),inv(14)(q11q32).</p>
Full article ">Figure 7
<p>(<b>a</b>) Maculopapular skin lesions on both lower extremities. (<b>b</b>) Skin biopsy (HE, 8× and insert 40×).</p>
Full article ">
17 pages, 1585 KiB  
Article
Disease-Modifying Therapies (DMTs) in Pregnant and Lactating Women with Multiple Sclerosis: Analysis of Real-World Data from EudraVigilance Database
by Liberata Sportiello, Raffaella Di Napoli, Nunzia Balzano, Annamaria Mascolo, Rosanna Ruggiero, Luigi Di Costanzo, Davida Monaco, Giorgia Teresa Maniscalco and Annalisa Capuano
Pharmaceuticals 2023, 16(11), 1566; https://doi.org/10.3390/ph16111566 - 6 Nov 2023
Viewed by 1402
Abstract
(1) Background: The purpose of study was to compare the safety profile of glatiramer with natalizumab, alemtuzumab and ocrelizumab in pregnant and lactating women affected by multiple sclerosis (MS). (2) Methods: Individual case safety reports (ICSRs) were retrieved from the European spontaneous reporting [...] Read more.
(1) Background: The purpose of study was to compare the safety profile of glatiramer with natalizumab, alemtuzumab and ocrelizumab in pregnant and lactating women affected by multiple sclerosis (MS). (2) Methods: Individual case safety reports (ICSRs) were retrieved from the European spontaneous reporting system database (EudraVigilance). The reporting odds ratios (RORs) were computed to compare the reporting probability of events between natalizumab, alemtuzumab and ocrelizumab vs. glatiramer. (3) Results: A total of 1236 ICSRs reporting at least one DMT as a suspected drug were selected. More adverse drug reactions (ADRs) unrelated to pregnancy and breastfeeding (n = 1171; 32.6%) were reported than ADRs specific to pregnancy and breastfeeding (n = 1093; 30.4%). The most frequently reported unrelated ADR was MS relapse. Alemtuzumab and natalizumab seem to have a lower reporting probability of MS relapse compared to glatiramer (ROR 0.17, 95% CI 0.07–0.45 and ROR 0.34, 95% CI 0.20–0.57). Among pregnancy- and breastfeeding-related ADRs, the first most reported event was spontaneous abortion (n = 321; 8.9%). Natalizumab and ocrelizumab were associated with a higher reporting probability of spontaneous abortion compared to glatiramer (ROR 2.22, 95% CI 1.58–3.12; ROR 2.18, 95% CI 1.34–3.54, respectively), while alemtuzumab had a lower reporting frequency (ROR 0.32, 95% CI 0.17–0.60). (4) Conclusions: This study did not suggest any strong or new insights for DMTs in this special subpopulation. However, further studies need to be performed. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

Figure 1
<p>Flowchart of the selection process of ICSRs from the EudraVigilance database.</p>
Full article ">Figure 2
<p>ROR of event groups for the comparisons between monoclonal antibodies ((<b>a</b>) alemtuzumab, (<b>b</b>) natalizumab and (<b>c</b>) ocrelizumab) and glatiramer. ROR, reporting odds ratio; 95% CI, 95% confidence interval.</p>
Full article ">Figure 3
<p>ROR of PTs “abortion spontaneous” (<b>a</b>) and “multiple sclerosis relapse” (<b>b</b>) for the comparisons between monoclonal antibodies (alemtuzumab, natalizumab and ocrelizumab) and glatiramer. ROR, reporting odds ratio; CI, confidence interval.</p>
Full article ">
20 pages, 3685 KiB  
Review
T-Cell Prolymphocytic Leukemia: Diagnosis, Pathogenesis, and Treatment
by Marc Gutierrez, Patrick Bladek, Busra Goksu, Carlos Murga-Zamalloa, Dale Bixby and Ryan Wilcox
Int. J. Mol. Sci. 2023, 24(15), 12106; https://doi.org/10.3390/ijms241512106 - 28 Jul 2023
Cited by 3 | Viewed by 5271
Abstract
T-cell prolymphocytic leukemia (T-PLL) is a rare and aggressive neoplasm of mature T-cells. Most patients with T-PLL present with lymphocytosis, anemia, thrombocytopenia, and hepatosplenomegaly. Correct identification of T-PLL is essential because treatment for this disease is distinct from that of other T-cell neoplasms. [...] Read more.
T-cell prolymphocytic leukemia (T-PLL) is a rare and aggressive neoplasm of mature T-cells. Most patients with T-PLL present with lymphocytosis, anemia, thrombocytopenia, and hepatosplenomegaly. Correct identification of T-PLL is essential because treatment for this disease is distinct from that of other T-cell neoplasms. In 2019, the T-PLL International Study Group (TPLL-ISG) established criteria for the diagnosis, staging, and assessment of response to treatment of T-PLL with the goal of harmonizing research efforts and supporting clinical decision-making. T-PLL pathogenesis is commonly driven by T-cell leukemia 1 (TCL1) overexpression and ATM loss, genetic alterations that are incorporated into the TPLL-ISG diagnostic criteria. The cooperativity between TCL1 family members and ATM is seemingly unique to T-PLL across the spectrum of T-cell neoplasms. The role of the T-cell receptor, its downstream kinases, and JAK/STAT signaling are also emerging themes in disease pathogenesis and have obvious therapeutic implications. Despite improved understanding of disease pathogenesis, alemtuzumab remains the frontline therapy in the treatment of naïve patients with indications for treatment given its high response rate. Unfortunately, the responses achieved are rarely durable, and the majority of patients are not candidates for consolidation with hematopoietic stem cell transplantation. Improved understanding of T-PLL pathogenesis has unveiled novel therapeutic vulnerabilities that may change the natural history of this lymphoproliferative neoplasm and will be the focus of this concise review. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Peripheral blood involvement by T-PLL. Representative images demonstrate that T-PLL tumor cells (Wright stain) can be variable in size and range from medium (<b>right panel</b>) to large (<b>left panel</b>), with irregular nuclear contours, condensed chromatin, and prominent nucleoli. The morphological and cytological features of T-PLL have been previously documented and described [<a href="#B2-ijms-24-12106" class="html-bibr">2</a>,<a href="#B6-ijms-24-12106" class="html-bibr">6</a>,<a href="#B7-ijms-24-12106" class="html-bibr">7</a>].</p>
Full article ">Figure 2
<p>(<b>A</b>,<b>B</b>) Involvement of bone marrow with T-PLL. (<b>A</b>) Highlights of interstitial involvement by tumor cells (blue arrows). (<b>B</b>) The most common morphological pattern is composed of medium lymphocytes with round nuclear contours and central prominent nucleoli (blue arrows). (<b>C</b>) Focal aggregates of tumor cells are usually observed (dotted circles). (<b>D</b>) Small-cell morphology variant is characterized by small to medium forms with condensed chromatin (blue arrows). Occasional large “cerebriform” lymphocytes are also observed (red arrows). The morphological features characteristic of T-PLL bone marrow involvement have been previously described [<a href="#B2-ijms-24-12106" class="html-bibr">2</a>,<a href="#B6-ijms-24-12106" class="html-bibr">6</a>,<a href="#B7-ijms-24-12106" class="html-bibr">7</a>].</p>
Full article ">Figure 3
<p>Skin involvement by T-PLL (black arrows). The tumor cells are predominantly distributed in a perivascular fashion, with no epidermotropism. Characteristic histological features of dermal involvement by T-PLL have been previously described [<a href="#B2-ijms-24-12106" class="html-bibr">2</a>,<a href="#B6-ijms-24-12106" class="html-bibr">6</a>,<a href="#B7-ijms-24-12106" class="html-bibr">7</a>].</p>
Full article ">Figure 4
<p>Therapeutic pathways in T-PLL. While alemtuzumab is the current frontline therapy, an improved understanding of T-PLL pathogenesis is leading to potentially targetable vulnerabilities. These include attempts to reinstate p53 function through MDM2 inhibitors as well as exploitation of faulty double strand break repair via PARP inhibitors, inhibition of TCR activation through ITK inhibitors as well as AKT/PI3K inhibitors, JAK inhibitors which inhibit cell growth and differentiation through the JAK-STAT pathway, BH3 mimetics to promote BAX and BAK mediated cell death, epigenetic approaches including hypomethylating agents and HDAC inhibitors, and novel antibody targeting e.g., CCR7. The illustration was created using Biorender.com and is adapted from previous review articles [<a href="#B47-ijms-24-12106" class="html-bibr">47</a>,<a href="#B72-ijms-24-12106" class="html-bibr">72</a>,<a href="#B73-ijms-24-12106" class="html-bibr">73</a>].</p>
Full article ">
13 pages, 457 KiB  
Review
Monoclonal Antibodies in Pregnancy and Breastfeeding in Patients with Multiple Sclerosis: A Review and an Updated Clinical Guide
by Panagiotis Gklinos and Ruth Dobson
Pharmaceuticals 2023, 16(5), 770; https://doi.org/10.3390/ph16050770 - 21 May 2023
Cited by 4 | Viewed by 4842
Abstract
The use of high-efficacy disease-modifying therapies (DMTs) early in the course of multiple sclerosis (MS) has been shown to improve clinical outcomes and is becoming an increasingly popular treatment strategy. As a result, monoclonal antibodies, including natalizumab, alemtuzumab, ocrelizumab, ofatumumab, and ublituximab, are [...] Read more.
The use of high-efficacy disease-modifying therapies (DMTs) early in the course of multiple sclerosis (MS) has been shown to improve clinical outcomes and is becoming an increasingly popular treatment strategy. As a result, monoclonal antibodies, including natalizumab, alemtuzumab, ocrelizumab, ofatumumab, and ublituximab, are frequently used for the treatment of MS in women of childbearing age. To date, only limited evidence is available on the use of these DMTs in pregnancy. We aim to provide an updated overview of the mechanisms of action, risks of exposure and treatment withdrawal, and pre-conception counseling and management during pregnancy and post-partum of monoclonal antibodies in women with MS. Discussing treatment options and family planning with women of childbearing age is essential before commencing a DMT in order to make the most suitable choice for each individual patient. Full article
(This article belongs to the Section Biopharmaceuticals)
Show Figures

Figure 1

Figure 1
<p>Summary of most important facts on the use of mAbs during pregnancy and breastfeeding. EID: extended interval dosing.</p>
Full article ">
11 pages, 1654 KiB  
Article
Targeted Large-Volume Lymphocyte Removal Using Magnetic Nanoparticles in Blood Samples of Patients with Chronic Lymphocytic Leukemia: A Proof-of-Concept Study
by Stefanie Janker, Simon Doswald, Roman R. Schimmer, Urs Schanz, Wendelin J. Stark, Martin Schläpfer and Beatrice Beck-Schimmer
Int. J. Mol. Sci. 2023, 24(8), 7523; https://doi.org/10.3390/ijms24087523 - 19 Apr 2023
Cited by 5 | Viewed by 1511
Abstract
In the past, our research group was able to successfully remove circulating tumor cells with magnetic nanoparticles. While these cancer cells are typically present in low numbers, we hypothesized that magnetic nanoparticles, besides catching single cells, are also capable of eliminating a large [...] Read more.
In the past, our research group was able to successfully remove circulating tumor cells with magnetic nanoparticles. While these cancer cells are typically present in low numbers, we hypothesized that magnetic nanoparticles, besides catching single cells, are also capable of eliminating a large number of tumor cells from the blood ex vivo. This approach was tested in a small pilot study in blood samples of patients suffering from chronic lymphocytic leukemia (CLL), a mature B-cell neoplasm. Cluster of differentiation (CD) 52 is a ubiquitously expressed surface antigen on mature lymphocytes. Alemtuzumab (MabCampath®) is a humanized, IgG1κ, monoclonal antibody directed against CD52, which was formerly clinically approved for treating chronic lymphocytic leukemia (CLL) and therefore regarded as an ideal candidate for further tests to develop new treatment options. Alemtuzumab was bound onto carbon-coated cobalt nanoparticles. The particles were added to blood samples of CLL patients and finally removed, ideally with bound B lymphocytes, using a magnetic column. Flow cytometry quantified lymphocyte counts before, after the first, and after the second flow across the column. A mixed effects analysis was performed to evaluate removal efficiency. p < 0.05 was defined as significant. In the first patient cohort (n = 10), using a fixed nanoparticle concentration, CD19-positive B lymphocytes were reduced by 38% and by 53% after the first and the second purification steps (p = 0.002 and p = 0.005), respectively. In a second patient cohort (n = 11), the nanoparticle concentration was increased, and CD19-positive B lymphocytes were reduced by 44% (p < 0.001) with no further removal after the second purification step. In patients with a high lymphocyte count (>20 G/L), an improved efficiency of approximately 20% was observed using higher nanoparticle concentrations. A 40 to 50% reduction of B lymphocyte count using alemtuzumab-coupled carbon-coated cobalt nanoparticles is feasible, also in patients with a high lymphocyte count. A second purification step did not further increase removal. This proof-of-concept study demonstrates that such particles allow for the targeted extraction of larger amounts of cellular blood components and might offer new treatment options in the far future. Full article
(This article belongs to the Special Issue Nanoparticles for Tumor Targeting and Therapy)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) shows the effect of alemtuzumab-nanoparticle treatment on CD19-positive B lymphocytes after 1 and 2 purification steps over a magnetic column. (<b>B</b>) In patients with a lower lymphocyte count (≤20 G/L blood), the purification appears to be more efficient than in (<b>C</b>) patients with a higher lymphocyte count. An example of gating is given in (<b>D</b>–<b>F</b>). Five thousand events were recorded using counting beads. The forward scatter (FSC-A) and phycoerythrin (PE-A) were plotted (<b>D</b>). Gating of the lymphocyte population was performed using the side scatter (SSC-A) and FSC-A (<b>E</b>) as described in the literature [<a href="#B14-ijms-24-07523" class="html-bibr">14</a>], followed by the exclusion of doubles by using forward scatter area (FSC-A) and forward scatter height (FSC-H). CD19+ B- and CD3+ T-lymphocyte populations were then gated using APC-A (bound to the CD19-antibody) and Alexa fluor 488 (bound to the CD3-antibody) (<b>F</b>).</p>
Full article ">Figure 2
<p>(<b>A</b>) shows the relative concentration of CD19-positive B lymphocytes to unpurified control after 1 and 2 alemtuzumab-nanoparticle purification treatments over a magnetic column. (<b>B</b>) The removal efficiency in patients with a lower lymphocyte count (≤20 G/L blood) was similar to the purification in patients with a (<b>C</b>) higher lymphocyte count despite an increase in nanoparticle concentration in these patients. An example of gating is given in D-F from a patient with more than 20 G lymphocytes/L. 5000 events were recorded using counting beads, gated with FSC-A and PE-A) (<b>D</b>). Gating of the lymphocyte population was performed using the side scatter (SSC-A) and FSC-A (<b>E</b>) as described in the literature [<a href="#B14-ijms-24-07523" class="html-bibr">14</a>], followed by the exclusion of doubles using FSC-A and FSC-H. CD19+ B- and CD3+ T-lymphocyte populations were then gated using APC-A (bound to the CD19-antibody) and Alexa fluor 488 (bound to the CD3-antibody) (<b>F</b>).</p>
Full article ">Figure 3
<p>(<b>A</b>) shows the average relative concentration of CD52 positive lymphocytes to unpurified control after 1 and 2 alemtuzumab-nanoparticle purification treatments. (<b>B</b>) The removal efficiency in patients with a lower lymphocyte count (≤20 G/L blood) was similar to the purification in patients with a (<b>C</b>) higher lymphocyte count despite doubling the nanoparticles in these patients. For the analysis, 5000 events were recorded using counting beads, gated with FSC-A and PE-A (<b>D</b>). Gating of the lymphocyte population was performed using the side scatter (SSC-A) and FSC-A (<b>E</b>) as described in the literature [<a href="#B14-ijms-24-07523" class="html-bibr">14</a>], followed by the exclusion of doubles using FSC-A and FSC-H. While a CD52 (marked with APC) and a CD3 (marked with Alexa Fluor 488) staining was performed, this experiment focused on the evaluation of CD52-positive cells irrespective of the CD3 staining (<b>F</b>).</p>
Full article ">
13 pages, 1038 KiB  
Article
Alemtuzumab-Related Lymphocyte Subset Dynamics and Disease Activity or Autoimmune Adverse Events: Real-World Evidence
by Elisabetta Signoriello, Giacomo Lus, Francesco Saccà, Marco Puthenparampil, Cinzia Coppola, Andrea Di Pietro, Gianfranco Puoti, Maria Cristina Criscuolo, Matteo Foschi, Giuseppina Miele, Gianmarco Abbadessa, Vincenzo Brescia Morra, Paolo Gallo, Simona Bonavita, Maria Pia Sormani and Alessio Signori
J. Clin. Med. 2023, 12(5), 1768; https://doi.org/10.3390/jcm12051768 - 22 Feb 2023
Cited by 2 | Viewed by 1684
Abstract
Background and objectives: alemtuzumab is a monoclonal anti-CD52 antibody acting on B and T cells in highly active multiple sclerosis (MS). We analyzed changes in lymphocyte subsets after alemtuzumab administration in relation to disease activity and autoimmune adverse events. Methods: lymphocyte subset counts [...] Read more.
Background and objectives: alemtuzumab is a monoclonal anti-CD52 antibody acting on B and T cells in highly active multiple sclerosis (MS). We analyzed changes in lymphocyte subsets after alemtuzumab administration in relation to disease activity and autoimmune adverse events. Methods: lymphocyte subset counts were assessed longitudinally using linear mixed models. Subset counts at baseline and during follow-up were correlated with relapse rate, adverse events, or magnetic resonance (MRI) activity. Results: we recruited 150 patients followed for a median of 2.7 years (IQR: 1.9–3.7). Total lymphocytes, CD4, CD8, and CD20 significantly decreased in all patients over 2 years (p < 0.001). Previous treatment with fingolimod increased the risk of disease activity and adverse events (p = 0.029). We found a higher probability of disease reactivation in males and in patients with over three active lesions at baseline. Higher EDSS scores at baseline and longer disease duration predicted the switch to other treatments after alemtuzumab. Discussion and conclusions: Our real-world study supports data from clinical trials in which lymphocyte subsets were not useful for predicting disease activity or autoimmune disease during treatment. The early use of an induction therapy such as alemtuzumab in patients with a lower EDSS score and short history of disease could mitigate the risk of treatment failure. Full article
(This article belongs to the Section Clinical Neurology)
Show Figures

Figure 1

Figure 1
<p>Lymphocyte dynamics in patients with and without clinical or instrumental activity. Clinical activity means the occurrence of disease relapses and instrumental activity means occurrence of gadolinium-enhancing lesions in MRI.</p>
Full article ">Figure 2
<p>Boxplot of lymphocyte subset longitudinal trends over 24 months. The line represents the longitudinal trend of the median value for each subset reported.</p>
Full article ">Figure 3
<p>Boxplot of lymphocyte subsets stratified for previous DMTs (fingolimod, natalizumab, or others) at baseline and at the last available observation within 24 months. Data at 24 months were used if available, otherwise 18 months or 12 months were considered.</p>
Full article ">
12 pages, 1719 KiB  
Article
Trans-Endothelial Migration of Memory T Cells Is Impaired in Alemtuzumab-Treated Multiple Sclerosis Patients
by Kristy Nguyen, Pierre Juillard, Simon Hawke, Georges E. Grau and Felix Marsh-Wakefield
J. Clin. Med. 2022, 11(21), 6266; https://doi.org/10.3390/jcm11216266 - 24 Oct 2022
Cited by 6 | Viewed by 1871
Abstract
The breakdown of the blood–brain barrier (BBB) and the trans-endothelial migration of lymphocytes are central events in the development of multiple sclerosis (MS). Autoreactive T cells are major players in MS pathogenesis, which are rapidly depleted following alemtuzumab treatment. This modulation, in turn, [...] Read more.
The breakdown of the blood–brain barrier (BBB) and the trans-endothelial migration of lymphocytes are central events in the development of multiple sclerosis (MS). Autoreactive T cells are major players in MS pathogenesis, which are rapidly depleted following alemtuzumab treatment. This modulation, in turn, inhibits CNS inflammation, but alemtuzumab’s effect on T cell migration into the CNS has been less studied. Human brain endothelial cells were stimulated with pro-inflammatory cytokines to mimic an inflamed BBB in vitro. Peripheral blood mononuclear cells from healthy controls, untreated or alemtuzumab-treated patients with relapsing-remitting MS (RRMS) were added to the BBB model to assess their transmigratory capacity. Here, the migration of CD4+ effector memory T (TEM) and CD8+ central memory T (TCM) cells across the BBB was impaired in alemtuzumab-treated patients. Naïve T (Tnaïve) cells were unable to migrate across all groups. CD38 was lowly expressed on CD8+ TCM cells, particularly for RRMS patients, compared to CD8+ Tnaïve cells. CD62L expression was lower on CD4+ TEM cells than CD4+ Tnaïve cells and decreased further in alemtuzumab-treated patients. These data suggest that repopulated memory T cells are phenotypically different from naïve T cells, which may affect their transmigration across the BBB in vitro. Full article
(This article belongs to the Special Issue Clinical Advances in Multiple Sclerosis)
Show Figures

Figure 1

Figure 1
<p><b>Alemtuzumab decreased the migratory capacity of CD4<sup>+</sup> T<sup>EM</sup> and CD8<sup>+</sup> T<sup>CM</sup> cells across the BBB in vitro.</b> PBMCs were isolated from healthy controls (n = 12, green squares), untreated RRMS (n = 6, blue triangles) and alemtuzumab-treated RRMS patients (n = 10, red circles). After stimulation of the HBEC monolayer, PBMCs were added to the transwell assay and were left overnight to migrate. Non-migrated and migrated cells were harvested from the BBB model and phenotyped for analysis as total cell numbers. (<b>A</b>) The number of (<b>i</b>) CD4<sup>+</sup> T<sup>EM</sup> cells and (<b>ii</b>) CD4<sup>+</sup> T<sup>naïve</sup> cells. (<b>B</b>) The number of (<b>i</b>) CD8<sup>high</sup> T<sup>CM</sup> cells and (<b>ii</b>) The CD8<sup>high</sup> T<sup>naïve</sup> cells. (<b>C</b>) The number of (<b>i</b>) CD8<sup>low</sup> T<sup>CM</sup> cells and (<b>ii</b>) CD8<sup>low</sup> T<sup>naïve</sup> cells. Wilcoxon matched-pairs signed-rank test. <span class="html-italic">p</span> ≤ 0.1 are shown. NM; non-migrating, M; migrating; T<sup>CM</sup>, central memory T cells; T<sup>EM</sup>, effector memory T cells; T<sup>naïve</sup>, naïve T cells; RRMS, relapsing-remitting multiple sclerosis.</p>
Full article ">Figure 2
<p><b>The mean fluorescence intensity (MFI) of CD38, CD49d and CD62L on fresh CD4<sup>+</sup> T<sup>EM</sup> and CD8<sup>+</sup> T<sup>CM</sup> cells compared to their naïve phenotypes.</b> The expression of cell surface markers was calculated in healthy controls (n = 12), untreated RRMS (n = 6) and alemtuzumab-treated RRMS patients (n = 10) for naïve (pink) and memory (blue) T cells. (<b>A</b>) CD38 expression on (<b>i</b>) CD4<sup>+</sup> T<sup>EM</sup> cells versus CD4<sup>+</sup> T<sup>naïve</sup> cells (<b>ii</b>) CD8<sup>high</sup> T<sup>CM</sup> cells versus CD8<sup>high</sup> T<sup>naïve</sup> cells (<b>iii</b>) CD8<sup>low</sup> T<sup>CM</sup> cells versus CD8<sup>low</sup> T<sup>naïve</sup> cells. (<b>B</b>) CD49d expression on (<b>i</b>) CD4<sup>+</sup> T<sup>EM</sup> cells versus CD4<sup>+</sup> T<sup>naïve</sup> cells (<b>ii</b>) CD8<sup>high</sup> T<sup>CM</sup> cells versus CD8<sup>high</sup> T<sup>naïve</sup> cells (<b>iii</b>) CD8<sup>low</sup> T<sup>CM</sup> cells versus CD8<sup>low</sup> T<sup>naïve</sup> cells. (<b>C</b>) CD62L expression on (<b>i</b>) CD4<sup>+</sup> T<sup>EM</sup> cells versus CD4<sup>+</sup> T<sup>naïve</sup> cells (<b>ii</b>) CD8<sup>high</sup> T<sup>CM</sup> cells versus CD8<sup>high</sup> T<sup>naïve</sup> cells (<b>iii</b>) CD8<sup>low</sup> T<sup>CM</sup> cells versus CD8<sup>low</sup> T<sup>naïve</sup> cells. Wilcoxon matched-paired signed-rank test compared paired naïve and memory T cells. A Kruskal–Wallis with Dunn’s multiple comparisons test was done to compare between groups. <span class="html-italic">p</span> ≤ 0.1 are shown. RRMS, relapsing-remitting multiple sclerosis; T<sup>CM</sup>, central memory T cells; T<sup>EM</sup>, effector memory T cells; T<sup>naïve</sup>, naïve T cells.</p>
Full article ">Figure 3
<p><b>Alemtuzumab depletes circulating T cell subsets.</b> Blood was sampled from healthy controls (n = 12, green squares), untreated RRMS (n = 6, blue triangles) and alemtuzumab-treated RRMS patients (n = 10, red circles). Upon sample collection, fresh PBMCs were immediately isolated and phenotyped. Each T cell subset was gated on to calculate the proportion of cells multiplied by the cell count and shown as cells per mL of blood. (<b>A</b>) The absolute number of (<b>i</b>) CD4<sup>+</sup> T<sup>EM</sup> cells, (<b>ii</b>) CD8<sup>high</sup> T<sup>CM</sup> cells and (<b>iii</b>) CD8<sup>low</sup> T<sup>CM</sup> cells. (<b>B</b>) The absolute number of <b>(i)</b> CD4<sup>+</sup> T<sup>naïve</sup> cells, (<b>ii</b>) CD8<sup>high</sup> T<sup>naïve</sup> cells and (<b>iii</b>) CD8<sup>low</sup> T<sup>naïve</sup> cells. Kruskal–Wallis with Dunn’s multiple comparisons test. <span class="html-italic">p</span> ≤ 0.1 are shown. T<sup>CM</sup>, central memory T cells; T<sup>EM</sup>, effector memory T cells; T<sup>naïve</sup>, naïve T cells. RRMS, relapsing-remitting multiple sclerosis.</p>
Full article ">
8 pages, 2282 KiB  
Article
Ella versus Simoa Serum Neurofilament Assessment to Monitor Treatment Response in Highly Active Multiple Sclerosis Patients
by Martin Nötzel, Luise Ina Werder, Tjalf Ziemssen and Katja Akgün
Int. J. Mol. Sci. 2022, 23(20), 12361; https://doi.org/10.3390/ijms232012361 - 15 Oct 2022
Cited by 9 | Viewed by 2691
Abstract
The measurement of serum neurofilament light chain (sNfL) is of growing importance in the field of neurology. In the management of multiple sclerosis, it can serve as a useful marker to assess disease activity and treatment response. This paper compares two available methods, [...] Read more.
The measurement of serum neurofilament light chain (sNfL) is of growing importance in the field of neurology. In the management of multiple sclerosis, it can serve as a useful marker to assess disease activity and treatment response. This paper compares two available methods, namely the Single Molecule Array (Simoa) and the Ella microfluid platform, to measure longitudinal sNfL levels of 42 highly active multiple sclerosis patients treated with alemtuzumab over a period of 36 months. In order to assess the methods agreement, Bland–Altman plots and Passing–Bablok regression were analyzed. Here, we show that despite the fact that Ella measures around 24% higher values than Simoa, both are equally suitable for longitudinal sNfL monitoring. Full article
(This article belongs to the Special Issue New Mechanisms and Therapeutics in Neurological Diseases 2.0)
Show Figures

Figure 1

Figure 1
<p>Comparison of Ella and Simoa methods. Bland–Altman plots in units (<b>A</b>) and percentage (<b>B</b>) with 95% confidence interval (CI) for mean, limit of agreement, and regression line. (<b>C</b>) Passing–Bablok regression with 95% CI. Dashed reference line is y = x, representing perfect method agreement.</p>
Full article ">Figure 2
<p>Timecourse of serum neurofilament light chain (sNfL) during alemtuzumab treatment in the whole cohort (<b>A</b>) in responders ((<b>B</b>), Months 15–36: No relapse activity, no MRI activity) and non-responders ((<b>C</b>), Months 15–36: Relapse activity and/or MRI activity), with 95% confidence intervals. The sNfL levels were measured using Ella and Simoa. (<b>A</b>: All patients (<span class="html-italic">n</span> = 42); (<b>B</b>) Responders (<span class="html-italic">n</span> = 29), (<b>C</b>): Non-responders (<span class="html-italic">n</span> = 13). Data time points are summarized quarterly.</p>
Full article ">
10 pages, 676 KiB  
Review
Adenovirus Infection in Pediatric Hematopoietic Cell Transplantation: A Challenge Still Open for Survival
by Simone Cesaro and Fulvio Porta
J. Clin. Med. 2022, 11(16), 4827; https://doi.org/10.3390/jcm11164827 - 18 Aug 2022
Cited by 15 | Viewed by 3272
Abstract
Human Adenovirus (HAdV) infection occurs in 14–16% of patients in the early months after pediatric hematopoietic cell transplantation (HCT) and this correlates with a higher risk of developing HAdV disease and overall 6-month mortality. The main risk factors for HAdV infection are T-cell [...] Read more.
Human Adenovirus (HAdV) infection occurs in 14–16% of patients in the early months after pediatric hematopoietic cell transplantation (HCT) and this correlates with a higher risk of developing HAdV disease and overall 6-month mortality. The main risk factors for HAdV infection are T-cell depletion of the graft by ex vivo CD34+ selection or in vivo use of alemtuzumab or anti-thymocyte serum, the development of grade III-IV graft versus host disease (GVHD), the type of donor (unrelated donor, cord blood, haploidentical, or HLA mismatched parent), and severe lymphopenia (<0.2 × 109/L). The prevention of HAdV disease is based on early intervention with antivirals in the asymptomatic patient when the permitted viral load threshold in the blood (≥102–3 copies/mL) and/or in the stool (109 copies/g stool) is exceeded. Cidofovir, a monophosphate nucleotide analog of cytosine, is the primary drug for preemptive therapy, used at 5 mg/kg/week for 2 weeks followed by 3–5 mg/kg every 2 weeks. The alternative schedule is 1 mg/kg every other day (three times/week). Enhancing virus-specific T-cell immunity in the first months post-HCT by donor-derived or third-party-derived virus-specific T cells represents an innovative and promising way of intervention, applicable both in prevention and therapeutic settings. Full article
(This article belongs to the Section Hematology)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of <span class="html-italic">Adenovirus</span> structure.</p>
Full article ">Figure 2
<p>Algorithm for monitoring and treatment of pediatric patients at high risk of adenovirus disease. Legend: ATG, anti-thymocyte globulin; HAdV, human adenovirus.</p>
Full article ">
11 pages, 1550 KiB  
Article
Decreased Intrathecal Concentrations of Free Light Chains Kappa in Multiple Sclerosis Patients Taking Very High Effective Disease-Modifying Treatment
by Marie Süße, Franz Felix Konen, Philipp Schwenkenbecher, Kathrin Budde, Matthias Nauck, Matthias Grothe, Malte Johannes Hannich and Thomas Skripuletz
Diagnostics 2022, 12(3), 720; https://doi.org/10.3390/diagnostics12030720 - 16 Mar 2022
Cited by 6 | Viewed by 1993
Abstract
Free light chains kappa (FLCκ) in cerebrospinal fluid (CSF) are a part of the intrathecal immune response. This observational study was conducted to investigate the effects of different disease-modifying therapies (DMT) on the humoral intrathecal immune response in the CSF of patients with [...] Read more.
Free light chains kappa (FLCκ) in cerebrospinal fluid (CSF) are a part of the intrathecal immune response. This observational study was conducted to investigate the effects of different disease-modifying therapies (DMT) on the humoral intrathecal immune response in the CSF of patients with multiple sclerosis (MS). FLCκ were analyzed in CSF and serum samples from MS patients taking DMT (n = 60) and those in a control cohort of treatment-naïve MS patients (n = 90). DMT was classified as moderately effective (including INFß-1a, INFß-1b, glatiramer acetate, dimethyl fumarate, teriflunomide, triamcinolone); highly effective (including fingolimod, daclizumab) and very highly effective (alemtuzumab, natalizumab, rituximab/ocrelizumab, mitoxantrone). FLCκ were measured using a nephelometric FLCκ kit. Intrathecal FLCκ and IgG concentrations were assessed in relation to the hyperbolic reference range in quotient diagrams. Intrathecal FLCκ concentrations and IgG concentrations were significantly lower in samples from the cohort of MS patients taking very highly effective DMT than in samples from the cohort of MS patients taking highly effective DMT and in the treatment-naïve cohort (FLCκ: p = 0.004, p < 0.0001 respectively/IgG: p = 0.013; p = 0.021). The reduction in FLCκ could contribute to an anti-inflammatory effect in the CNS through this mechanism. There was no difference in the appearance of CSF-specific oligoclonal bands (p = 0.830). Longitudinal analyses are required to confirm these results. Full article
(This article belongs to the Section Pathology and Molecular Diagnostics)
Show Figures

Figure 1

Figure 1
<p>Data on local IgG concentrations of patient samples presented in box plots (median, first, and third quartile). Samples from patients treated with very highly effective DMT had significantly lower IgG concentrations than samples treated with highly effective DMT (<span class="html-italic">p</span> = 0.013) and the control group (<span class="html-italic">p</span> = 0.021). DMT—disease-modifying therapy, Ig—Immunoglobulin. Comparison between groups was performed using the Kruskal–Wallis test. The Dunn–Bonferroni test was used for post hoc analysis.</p>
Full article ">Figure 2
<p>Data of local FLCκ concentrations of patient samples presented in box plots (median, first, and third quartile). Samples from patients treated with very high effective DMT had significantly lower FLCκ concentrations than samples treated with high effective DMT (<span class="html-italic">p</span> = 0.004) and the control group (<span class="html-italic">p</span> &lt; 0.0001). DMT—disease-modifying therapy, FLCκ—free light chains kappa. Comparison between groups was performed using the Kruskal–Wallis test. The Dunn–Bonferroni test was used for post hoc analysis.</p>
Full article ">Figure 3
<p>Local FLCκ and IgG concentration of patient samples distributed after ingestion of DMT (median, first, and third quartile)<b>.</b> FLCκ—free light chains kappa, DMT—disease-modifying therapy, INF—interferon, GLAT—glatiramer acetate.</p>
Full article ">
13 pages, 335 KiB  
Article
Validation of De Novo Peptide Sequences with Bottom-Up Tag Convolution
by Kira Vyatkina
Proteomes 2022, 10(1), 1; https://doi.org/10.3390/proteomes10010001 - 29 Dec 2021
Cited by 2 | Viewed by 2848
Abstract
De novo sequencing is indispensable for the analysis of proteins from organisms with unknown genomes, novel splice variants, and antibodies. However, despite a variety of methods developed to this end, distinguishing between the correct interpretation of a mass spectrum and a number of [...] Read more.
De novo sequencing is indispensable for the analysis of proteins from organisms with unknown genomes, novel splice variants, and antibodies. However, despite a variety of methods developed to this end, distinguishing between the correct interpretation of a mass spectrum and a number of incorrect alternatives often remains a challenge. Tag convolution is computed for a set of peptide sequence tags of a fixed length k generated from the input tandem mass spectra and can be viewed as a generalization of the well-known spectral convolution. We demonstrate its utility for validating de novo peptide sequences by using a set of those generated by the algorithm PepNovo+ from high-resolution bottom-up data sets for carbonic anhydrase 2 and the Fab region of alemtuzumab and indicate its further potential applications. Full article
Show Figures

Figure 1

Figure 1
<p>For a toy input set <math display="inline"><semantics> <mrow> <mi mathvariant="script">S</mi> <mo>=</mo> <mo>{</mo> <msub> <mi>S</mi> <mn>1</mn> </msub> <mo>,</mo> <msub> <mi>S</mi> <mn>2</mn> </msub> <mo>,</mo> <msub> <mi>S</mi> <mn>3</mn> </msub> <mo>}</mo> </mrow> </semantics></math>, spectra <math display="inline"><semantics> <msub> <mi>S</mi> <mn>1</mn> </msub> </semantics></math>, <math display="inline"><semantics> <msub> <mi>S</mi> <mn>2</mn> </msub> </semantics></math>, and <math display="inline"><semantics> <msub> <mi>S</mi> <mn>3</mn> </msub> </semantics></math> contain one, two, and one 3-tag(s), respectively. Here, <math display="inline"><semantics> <mrow> <mi>τ</mi> <mo>(</mo> <mi>A</mi> <mi>C</mi> <mi>D</mi> <mo>,</mo> <mi>E</mi> <mi>F</mi> <mi>G</mi> <mo>)</mo> <mo>=</mo> <mo>{</mo> <mo>(</mo> <mn>500</mn> <mo>,</mo> <mn>2</mn> <mo>)</mo> <mo>,</mo> <mo>(</mo> <mn>700</mn> <mo>,</mo> <mn>1</mn> <mo>)</mo> <mo>}</mo> </mrow> </semantics></math>.</p>
Full article ">Figure 2
<p>Four spectra acquired from a toy protein with the amino acid sequence AVTDPVLSGNATSMPGST. Tag convolution is being computed for the strings TDPVL and ATSMP. Two tags composing a pair that contributes the “correct” (i.e., equal to <math display="inline"><semantics> <mrow> <mi>m</mi> <mo>(</mo> <mi>S</mi> <mi>G</mi> <mi>N</mi> <mo>)</mo> </mrow> </semantics></math>) value can be derived from the following: (<b>a</b>) a spectrum acquired from the entire protein; (<b>b</b>) a spectrum acquired from a fragment of the underlying protein; (<b>c</b>) two distinct spectra acquired from possibly different protein fragments starting at a same amino acid residue.</p>
Full article ">Figure 3
<p>Four spectra acquired from a toy protein with the amino acid sequence <span class="html-italic">s</span> = AVTDPVLSGNATSMPGST together give rise to five 3-tags. The pairs of tags labeled with DPV and SMP, PVL and SMP, STA and VPD, and STA and GSL, respectively, contribute to the tag and 3-mer scores of certain amino acids of <span class="html-italic">s</span>.</p>
Full article ">Figure 4
<p>Contribution of a pair of 3-mers to <math display="inline"><semantics> <mrow> <mi>T</mi> <mo>(</mo> <mrow> <mi>AVTDPLSG</mi> </mrow> <mo>,</mo> <mrow> <mi>ATSMPGST</mi> </mrow> <mo>)</mo> </mrow> </semantics></math> computed at the time of validating the amino acid N-10 of the toy protein from <a href="#proteomes-10-00001-f002" class="html-fig">Figure 2</a>. (<b>a</b>) The protein sequence. (<b>b</b>) To validate N-10, we consider the prefix <math display="inline"><semantics> <msub> <mi>s</mi> <mn>1</mn> </msub> </semantics></math> and suffix <math display="inline"><semantics> <msub> <mi>s</mi> <mn>2</mn> </msub> </semantics></math> of the entire sequence immediately preceding and following N-10, respectively, and examine all the pairs of 3-mers from <math display="inline"><semantics> <msub> <mi>s</mi> <mn>1</mn> </msub> </semantics></math> and <math display="inline"><semantics> <msub> <mi>s</mi> <mn>2</mn> </msub> </semantics></math>, respectively, and from <math display="inline"><semantics> <mover> <msub> <mi>s</mi> <mn>2</mn> </msub> <mo>¯</mo> </mover> </semantics></math> and <math display="inline"><semantics> <mover> <msub> <mi>s</mi> <mn>1</mn> </msub> <mo>¯</mo> </mover> </semantics></math>, respectively. (<b>c</b>) Processing of the pair of 3-mers <math display="inline"><semantics> <msub> <mi>w</mi> <mn>1</mn> </msub> </semantics></math> = DPV and <math display="inline"><semantics> <msub> <mi>w</mi> <mn>2</mn> </msub> </semantics></math> = SMP from <math display="inline"><semantics> <msub> <mi>s</mi> <mn>1</mn> </msub> </semantics></math> and <math display="inline"><semantics> <msub> <mi>s</mi> <mn>2</mn> </msub> </semantics></math>, respectively. To either 3-mer, precisely one 3-tag from the set <math display="inline"><semantics> <mi mathvariant="script">T</mi> </semantics></math> depicted in <a href="#proteomes-10-00001-f003" class="html-fig">Figure 3</a> corresponds. Both tags are defined by <span class="html-italic">b</span>-ions and properly align against the sequence. Thus, the difference between their offsets, which contributes to <math display="inline"><semantics> <mrow> <mi>τ</mi> <mo>(</mo> <msub> <mi>w</mi> <mn>1</mn> </msub> <mo>,</mo> <msub> <mi>w</mi> <mn>2</mn> </msub> <mo>)</mo> </mrow> </semantics></math>, equals <math display="inline"><semantics> <mrow> <mi>M</mi> <mi>a</mi> <mi>s</mi> <mi>s</mi> <mo>(</mo> <mrow> <mi>DPVLSGNAT</mi> </mrow> <mo>)</mo> </mrow> </semantics></math>. When shifting this value by <math display="inline"><semantics> <mrow> <mi>δ</mi> <mo>=</mo> <mo>−</mo> <mi>M</mi> <mi>a</mi> <mi>s</mi> <mi>s</mi> <mo>(</mo> <mrow> <mi>DPVLSG</mi> </mrow> <mo>)</mo> <mo>−</mo> <mi>M</mi> <mi>a</mi> <mi>s</mi> <mi>s</mi> <mo>(</mo> <mrow> <mi>AT</mi> </mrow> <mo>)</mo> </mrow> </semantics></math>, we obtain the mass of N equal to 114. Consequently, the pair (DPV,SMP) of 3-mers contributes to <math display="inline"><semantics> <mrow> <mi>T</mi> <mo>(</mo> <mrow> <mi>AVTDPLSG</mi> </mrow> <mo>,</mo> <mrow> <mi>ATSMPGST</mi> </mrow> <mo>)</mo> </mrow> </semantics></math> with a value of 114 with multiplicity 1.</p>
Full article ">
20 pages, 862 KiB  
Review
Targeting Antigen-Presenting Cells in Multiple Sclerosis Treatment
by Piotr Szpakowski, Dominika Ksiazek-Winiarek and Andrzej Glabinski
Appl. Sci. 2021, 11(18), 8557; https://doi.org/10.3390/app11188557 - 15 Sep 2021
Cited by 3 | Viewed by 2729
Abstract
Multiple sclerosis (MS) is common neurological disease of the central nervous system (CNS) affecting mostly young adults. Despite decades of studies, its etiology and pathogenesis are not fully unraveled and treatment is still insufficient. The vast majority of studies suggest that the immune [...] Read more.
Multiple sclerosis (MS) is common neurological disease of the central nervous system (CNS) affecting mostly young adults. Despite decades of studies, its etiology and pathogenesis are not fully unraveled and treatment is still insufficient. The vast majority of studies suggest that the immune system plays a major role in MS development. This is also supported by the effectiveness of currently available MS treatments that target immunocompetent cells. In this review, the role of antigen-presenting cells (APC) in MS development as well as the novel therapeutic options targeting those cells in MS are presented. It is known that in MS, peripheral self-antigen-specific immune cells are activated during antigen presentation process and they enter the CNS through the disrupted blood–brain barrier (BBB). Myelin-reactive CD4+ T-cells can be activated by dendritic cells, infiltrating macrophages, microglia cells, or B-cells, which all express MHC class II molecules. There are also suggestions that brain endothelial cells may act as non-professional APCs and present myelin-specific antigens with MHC class II. Similarly, astrocytes, the major glial cells in the CNS, were shown to act as non-professional APCs presenting myelin antigens to autoreactive T-cells. Several currently available MS drugs such as natalizumab, fingolimod, alemtuzumab, and ocrelizumab may modulate antigen presentation in MS. Another way to use this mechanism in MS treatment may be the usage of specific tolerogenic dendritic cells or the induction of tolerance to myelin antigens by peptide vaccines. Full article
(This article belongs to the Section Applied Biosciences and Bioengineering)
Show Figures

Figure 1

Figure 1
<p>Clinical studies conducted on tolerogenic dendritic cells.</p>
Full article ">
Back to TopTop