1
|
Looney CM, Strauli N, Cascino MD, Garma H, Schroeder AV, Takahashi C, O'Gorman W, Green C, Herman AE. Development of a novel, highly sensitive assay for quantification of minimal residual B cells in autoimmune disease and comparison to traditional methods across B-cell-depleting agents. Clin Immunol 2023; 248:109265. [PMID: 36796471 DOI: 10.1016/j.clim.2023.109265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023]
Abstract
Targeted B-cell depletion is a useful therapy for many diseases, including autoimmune disorders and certain cancers. We developed a sensitive blood B-cell depletion assay, MRB 1.1, compared its performance with the T-cell/B-cell/NK-cell (TBNK) assay, and assessed B-cell depletion with different therapies. The empirically defined lower limit of quantification (LLOQ) for CD19+ cells in the TBNK assay was 10 cells/μL, and 0.441 cells/μL for the MRB 1.1 assay. The TBNK LLOQ was used to compare differences between B-cell depletion in similar lupus nephritis patient populations who received rituximab (LUNAR), ocrelizumab (BELONG), or obinutuzumab (NOBILITY). After 4 weeks, 10% of patients treated with rituximab retained detectable B cells vs 1.8% with ocrelizumab and 1.7% for obinutuzumab; at 24 weeks 93% of patients who received obinutuzumab remained below LLOQ vs 63% for rituximab. More-sensitive measurements of B cells may reveal differences in potency among anti-CD20 agents, which may associate with clinical outcomes.
Collapse
|
2
|
Hauser SL, Bar-Or A, Weber MS, Kletzl H, Günther A, Manfrini M, Model F, Mercier F, Petry C, Wing Q, Koendgen H, Smith T, Kappos L. Association of Higher Ocrelizumab Exposure With Reduced Disability Progression in Multiple Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:10/2/e200094. [PMID: 36792367 PMCID: PMC9931184 DOI: 10.1212/nxi.0000000000200094] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 12/19/2022] [Indexed: 02/17/2023]
Abstract
BACKGROUND AND OBJECTIVES Ocrelizumab improved clinical and MRI measures of disease activity and progression in three phase 3 multiple sclerosis (MS) studies. Post hoc analyses demonstrated a correlation between the ocrelizumab serum concentration and the degree of blood B-cell depletion, and body weight was identified as the most influential covariate on ocrelizumab pharmacokinetics. The magnitude of ocrelizumab treatment benefit on disability progression was greater in lighter vs heavier patients. These observations suggest that higher ocrelizumab serum levels provide more complete B-cell depletion and a greater delay in disability progression. The current post hoc analyses assessed population exposure-efficacy/safety relationships of ocrelizumab in patients with relapsing and primary progressive MS. METHODS Patients in OPERA I/II and ORATORIO were grouped in exposure quartiles based on their observed individual serum ocrelizumab level over the treatment period. Exposure-response relationships were analyzed for clinical efficacy (24-week confirmed disability progression (CDP), annualized relapse rate [ARR], and MRI outcomes) and adverse events. RESULTS Ocrelizumab reduced new MRI lesion counts to nearly undetectable levels in patients with relapsing or primary progressive MS across all exposure subgroups, and reduced ARR in patients with relapsing MS to very low levels (0.13-0.18). A consistent trend of higher ocrelizumab exposure leading to lower rates of CDP was seen (0%-25% [lowest] to 75%-100% [highest] quartile hazard ratios and 95% confidence intervals; relapsing MS: 0.70 [0.41-1.19], 0.85 [0.52-1.39], 0.47 [0.25-0.87], and 0.34 [0.17-0.70] vs interferon β-1a; primary progressive MS: 0.88 [0.59-1.30], 0.86 [0.60-1.25], 0.77 [0.52-1.14], and 0.55 [0.36-0.83] vs placebo). Infusion-related reactions, serious adverse events, and serious infections were similar across exposure subgroups. DISCUSSION The almost complete reduction of ARR and MRI activity already evident in the lowest quartile, and across all ocrelizumab-exposure groups, suggests a ceiling effect. A consistent trend of higher ocrelizumab exposure leading to greater reduction in risk of CDP was observed, particularly in the relapsing MS trials, and was not associated with a higher rate of adverse events. Higher ocrelizumab exposure may provide improved control of disability progression by reducing disease activity below that detectable by ARR and MRI, and/or by attenuating other B-cell-related pathologies responsible for tissue damage. CLASSIFICATION OF EVIDENCE This analysis provides Class III evidence that higher ocrelizumab serum levels are related to greater reduction in risk of disability progression in patients with multiple sclerosis. The study is rated Class III because of the initial treatment randomization disclosure that occurred after inclusion in the open-label extension. TRIAL REGISTRATION INFORMATION ClinicalTrials.gov Identifier: NCT01247324 (OPERA I), NCT01412333 (OPERA II), and NCT01194570 (ORATORIO).
Collapse
Affiliation(s)
- Stephen L Hauser
- From the UCSF Weill Institute for Neurosciences and Department of Neurology, University of California, San Francisco (S.L.H.); Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute of Neuropathology and Department of Neurology (M.S.W.), Universitätsmedizin Göttingen Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany; F. Hoffmann-La Roche Ltd (H. Kletzl, A.G., M.M., F. Model, F. Mercier, C.P., Q.W., H. Koendgen), Basel, Switzerland; NeuMatRx Ltd (T.S.), Bath, UK; and University Hospital Basel (L.K.), University of Basel, Switzerland.
| | - Amit Bar-Or
- From the UCSF Weill Institute for Neurosciences and Department of Neurology, University of California, San Francisco (S.L.H.); Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute of Neuropathology and Department of Neurology (M.S.W.), Universitätsmedizin Göttingen Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany; F. Hoffmann-La Roche Ltd (H. Kletzl, A.G., M.M., F. Model, F. Mercier, C.P., Q.W., H. Koendgen), Basel, Switzerland; NeuMatRx Ltd (T.S.), Bath, UK; and University Hospital Basel (L.K.), University of Basel, Switzerland
| | - Martin S Weber
- From the UCSF Weill Institute for Neurosciences and Department of Neurology, University of California, San Francisco (S.L.H.); Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute of Neuropathology and Department of Neurology (M.S.W.), Universitätsmedizin Göttingen Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany; F. Hoffmann-La Roche Ltd (H. Kletzl, A.G., M.M., F. Model, F. Mercier, C.P., Q.W., H. Koendgen), Basel, Switzerland; NeuMatRx Ltd (T.S.), Bath, UK; and University Hospital Basel (L.K.), University of Basel, Switzerland
| | - Heidemarie Kletzl
- From the UCSF Weill Institute for Neurosciences and Department of Neurology, University of California, San Francisco (S.L.H.); Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute of Neuropathology and Department of Neurology (M.S.W.), Universitätsmedizin Göttingen Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany; F. Hoffmann-La Roche Ltd (H. Kletzl, A.G., M.M., F. Model, F. Mercier, C.P., Q.W., H. Koendgen), Basel, Switzerland; NeuMatRx Ltd (T.S.), Bath, UK; and University Hospital Basel (L.K.), University of Basel, Switzerland
| | - Andreas Günther
- From the UCSF Weill Institute for Neurosciences and Department of Neurology, University of California, San Francisco (S.L.H.); Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute of Neuropathology and Department of Neurology (M.S.W.), Universitätsmedizin Göttingen Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany; F. Hoffmann-La Roche Ltd (H. Kletzl, A.G., M.M., F. Model, F. Mercier, C.P., Q.W., H. Koendgen), Basel, Switzerland; NeuMatRx Ltd (T.S.), Bath, UK; and University Hospital Basel (L.K.), University of Basel, Switzerland
| | - Marianna Manfrini
- From the UCSF Weill Institute for Neurosciences and Department of Neurology, University of California, San Francisco (S.L.H.); Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute of Neuropathology and Department of Neurology (M.S.W.), Universitätsmedizin Göttingen Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany; F. Hoffmann-La Roche Ltd (H. Kletzl, A.G., M.M., F. Model, F. Mercier, C.P., Q.W., H. Koendgen), Basel, Switzerland; NeuMatRx Ltd (T.S.), Bath, UK; and University Hospital Basel (L.K.), University of Basel, Switzerland
| | - Fabian Model
- From the UCSF Weill Institute for Neurosciences and Department of Neurology, University of California, San Francisco (S.L.H.); Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute of Neuropathology and Department of Neurology (M.S.W.), Universitätsmedizin Göttingen Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany; F. Hoffmann-La Roche Ltd (H. Kletzl, A.G., M.M., F. Model, F. Mercier, C.P., Q.W., H. Koendgen), Basel, Switzerland; NeuMatRx Ltd (T.S.), Bath, UK; and University Hospital Basel (L.K.), University of Basel, Switzerland
| | - Francois Mercier
- From the UCSF Weill Institute for Neurosciences and Department of Neurology, University of California, San Francisco (S.L.H.); Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute of Neuropathology and Department of Neurology (M.S.W.), Universitätsmedizin Göttingen Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany; F. Hoffmann-La Roche Ltd (H. Kletzl, A.G., M.M., F. Model, F. Mercier, C.P., Q.W., H. Koendgen), Basel, Switzerland; NeuMatRx Ltd (T.S.), Bath, UK; and University Hospital Basel (L.K.), University of Basel, Switzerland
| | - Claire Petry
- From the UCSF Weill Institute for Neurosciences and Department of Neurology, University of California, San Francisco (S.L.H.); Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute of Neuropathology and Department of Neurology (M.S.W.), Universitätsmedizin Göttingen Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany; F. Hoffmann-La Roche Ltd (H. Kletzl, A.G., M.M., F. Model, F. Mercier, C.P., Q.W., H. Koendgen), Basel, Switzerland; NeuMatRx Ltd (T.S.), Bath, UK; and University Hospital Basel (L.K.), University of Basel, Switzerland
| | - Qing Wing
- From the UCSF Weill Institute for Neurosciences and Department of Neurology, University of California, San Francisco (S.L.H.); Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute of Neuropathology and Department of Neurology (M.S.W.), Universitätsmedizin Göttingen Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany; F. Hoffmann-La Roche Ltd (H. Kletzl, A.G., M.M., F. Model, F. Mercier, C.P., Q.W., H. Koendgen), Basel, Switzerland; NeuMatRx Ltd (T.S.), Bath, UK; and University Hospital Basel (L.K.), University of Basel, Switzerland
| | - Harold Koendgen
- From the UCSF Weill Institute for Neurosciences and Department of Neurology, University of California, San Francisco (S.L.H.); Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute of Neuropathology and Department of Neurology (M.S.W.), Universitätsmedizin Göttingen Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany; F. Hoffmann-La Roche Ltd (H. Kletzl, A.G., M.M., F. Model, F. Mercier, C.P., Q.W., H. Koendgen), Basel, Switzerland; NeuMatRx Ltd (T.S.), Bath, UK; and University Hospital Basel (L.K.), University of Basel, Switzerland
| | - Terence Smith
- From the UCSF Weill Institute for Neurosciences and Department of Neurology, University of California, San Francisco (S.L.H.); Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute of Neuropathology and Department of Neurology (M.S.W.), Universitätsmedizin Göttingen Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany; F. Hoffmann-La Roche Ltd (H. Kletzl, A.G., M.M., F. Model, F. Mercier, C.P., Q.W., H. Koendgen), Basel, Switzerland; NeuMatRx Ltd (T.S.), Bath, UK; and University Hospital Basel (L.K.), University of Basel, Switzerland
| | - Ludwig Kappos
- From the UCSF Weill Institute for Neurosciences and Department of Neurology, University of California, San Francisco (S.L.H.); Department of Neurology and Center for Neuroinflammation and Experimental Therapeutics (A.B.-O.), Perelman School of Medicine, University of Pennsylvania, Philadelphia; Institute of Neuropathology and Department of Neurology (M.S.W.), Universitätsmedizin Göttingen Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, Göttingen, Germany; F. Hoffmann-La Roche Ltd (H. Kletzl, A.G., M.M., F. Model, F. Mercier, C.P., Q.W., H. Koendgen), Basel, Switzerland; NeuMatRx Ltd (T.S.), Bath, UK; and University Hospital Basel (L.K.), University of Basel, Switzerland
| |
Collapse
|
3
|
de Jong W, Aerts J, Allard S, Brander C, Buyze J, Florence E, van Gorp E, Vanham G, Leal L, Mothe B, Thielemans K, Plana M, Garcia F, Gruters R. iHIVARNA phase IIa, a randomized, placebo-controlled, double-blinded trial to evaluate the safety and immunogenicity of iHIVARNA-01 in chronically HIV-infected patients under stable combined antiretroviral therapy. Trials 2019; 20:361. [PMID: 31208472 PMCID: PMC6580477 DOI: 10.1186/s13063-019-3409-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 05/06/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND HIV therapeutic vaccination aims to improve the immune responses against HIV in order to control viral replication without the need for combined antiretroviral therapy (cART). iHIVARNA-01 is a novel vaccine combining mRNA delivery and T-cell immunogen (HTI) based on conserved targets of effective antiviral T-cell responses. In addition, it holds adequate stimuli required for activating antigen presenting cells (APC)s and co-activating specific T-cells (TriMix), including human CD40L, constitutively active TLR4 (caTLR4) and CD70. We propose that in-vivo targeting of dendritic cells (DCs) by direct administration of a HIV mRNA encoding these immune modulating proteins might be an attractive alternative to target DCs in vitro. METHODS/DESIGN This is a phase-IIa, randomized, double-blinded, placebo-controlled, multicenter study in chronically HIV-1 infected patients under stable cART. One of the three study arms is randomly allocated to subjects. Three vaccinations with either HIVACAT T-cell immunogen (HTI)-TriMix (iHIVARNA-01), TriMix or water for injection (WFI) (weeks 0, 2 and 4) are administered by intranodal injection in the inguinal region. Two weeks after the last immunization (week 6) cART is stopped for 12 weeks. The two primary endpoints are: (1) safety and tolerability of intranodal iHIVARNA-01 vaccination compared with TriMix or WFI and (2) induced immunogenicity, i.e., increase in the frequency of HIV-specific T-cell responses between baseline, week 6 and 12 weeks after treatment interruption in iHIVARNA-01-treated patients as compared to the control groups, immunized with TriMix-mRNA or WFI measured by an IFNγ ELISPOT assay. Secondary endpoints include the evaluation of time to viral rebound, plasma viral load (pVL) at w18, the proportion of patients with control of viral load, induction of T-cell responses to new HIV epitopes, polyfunctionality of HIV-specific T-cells, CD8+ T-cell in-vitro HIV suppressive capacity, the effect on viral reservoir (measured by proviral DNA and cell-associated RNA), assessment of viral immune escape by mutation and mRNA expression profiles of host immune genes. DISCUSSION This trial aims to direct target DC in situ with mRNA encoding HTI and TriMix for co-stimulation. Intranodal injection circumvents laborious DC isolation and handling in the laboratory. The trial extends on the safety results of a phase-I dose-escalating trial. This candidate vaccine could complement or even replace cART for chronic HIV infection and could be applicable to improve the care and cost of HIV infection. TRIAL REGISTRATION EudraCT 2016-002724-83 (22 September 2016); ClinicalTrials.gov, ID: NCT02888756 . Registered on 23 August 2016.
Collapse
Affiliation(s)
- Wesley de Jong
- Department of Viroscience, Erasmus MC, Room Ee-1726, P.O. Box 2040, 3000, CA, Rotterdam, The Netherlands
| | - Joeri Aerts
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sabine Allard
- Department of Internal Medicine and Infectious Diseases, Universitair Ziekenhuis Brussel, Brussels, Belgium
| | - Christian Brander
- Infectious Diseases Unit, IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Badalona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain.,AELIX Therapeutics, Parc Científic de Barcelona, Barcelona, Spain.,University of Vic - Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Jozefien Buyze
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine and, Antwerp, Belgium.,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Eric Florence
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine and, Antwerp, Belgium.,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Eric van Gorp
- Department of Viroscience, Erasmus MC, Room Ee-1726, P.O. Box 2040, 3000, CA, Rotterdam, The Netherlands
| | - Guido Vanham
- Virology Unit, Department of Biomedical Sciences, Institute of Tropical Medicine and, Antwerp, Belgium.,Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Lorna Leal
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Villarroel, 170, 08036, Barcelona, Spain.,Infectious Diseases Unit, Hospital Clínic, Villarroel, 170, 08036, Barcelona, Spain
| | - Beatriz Mothe
- Infectious Diseases Unit, IrsiCaixa AIDS Research Institute, Hospital Germans Trias i Pujol, Badalona, Spain.,University of Vic - Central University of Catalonia (UVic-UCC), Vic, Spain
| | - Kris Thielemans
- Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel, Brussels, Belgium
| | - Montse Plana
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Villarroel, 170, 08036, Barcelona, Spain
| | - Félipe Garcia
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Villarroel, 170, 08036, Barcelona, Spain. .,Infectious Diseases Unit, Hospital Clínic, Villarroel, 170, 08036, Barcelona, Spain.
| | - Rob Gruters
- Department of Viroscience, Erasmus MC, Room Ee-1726, P.O. Box 2040, 3000, CA, Rotterdam, The Netherlands.
| | | |
Collapse
|
4
|
Ondigo BN, Ndombi EM, Nicholson SC, Oguso JK, Carter JM, Kittur N, Secor WE, Karanja DMS, Colley DG. Functional Studies of T Regulatory Lymphocytes in Human Schistosomiasis in Western Kenya. Am J Trop Med Hyg 2018; 98:1770-1781. [PMID: 29692308 PMCID: PMC6086154 DOI: 10.4269/ajtmh.17-0966] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Immunoregulation is considered a common feature of Schistosoma mansoni infections, and elevated levels of T regulatory (Treg) lymphocytes have been reported during chronic human schistosomiasis. We now report that the removal of Treg (CD4+/CD25hi/CD127low lymphocytes) from peripheral blood mononuclear cells (PBMCs) of S. mansoni–infected individuals leads to increased levels of phytohemagglutinin (PHA)-stimulated interferon gamma (IFNγ) production and decreased interleukin-10 (IL-10) responses. Exposure to schistosome antigens did not result in measurable IFNγ by either PBMC or Treg-depleted populations. Interleukin-10 responses to soluble egg antigens (SEA) by PBMC were unchanged by Treg depletion, but the depletion of Treg greatly decreased IL-10 production to soluble worm antigenic preparation (SWAP). Proliferative responses to PHA increased upon Treg removal, but responses to SEA or SWAP did not, unless only initially low responders were evaluated. Addition of anti-IL-10 increased PBMC proliferative responses to either SEA or SWAP, but did not alter responses by Treg-depleted cells. Blockade by anti-transforming growth factor-beta (TGF-β) increased SEA but not SWAP proliferative responses by PBMC, whereas anti-TGF-β increased both SEA- and SWAP-stimulated responses by Treg-depleted cultures. Addition of both anti-IL-10 and anti-TGF-β to PBMC or Treg-depleted populations increased proliferation of both populations to either SEA or SWAP. These studies demonstrate that Treg appear to produce much of the antigen-stimulated IL-10, but other cell types or subsets of Treg may produce much of the TGF-β. The elevated levels of Treg seen in chronic schistosomiasis appear functional and involve IL-10 and TGF-β in antigen-specific immunoregulation perhaps leading to regulation of immunopathology and/or possibly decreased immunoprotective responses.
Collapse
Affiliation(s)
- Bartholomew N Ondigo
- Department of Biochemistry and Molecular Biology, Egerton University, Nakuru, Kenya.,Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Eric M Ndombi
- Department of Pathology, Kenyatta University, Nairobi, Kenya.,Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Sarah C Nicholson
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia
| | - John K Oguso
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Jennifer M Carter
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia
| | - Nupur Kittur
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia
| | - W Evan Secor
- Division of Parasitic Diseases and Malaria, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Diana M S Karanja
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Daniel G Colley
- Center for Tropical and Emerging Global Diseases, University of Georgia, Athens, Georgia.,Department of Microbiology, University of Georgia, Athens, Georgia
| |
Collapse
|
5
|
Greenfield AL, Hauser SL. B-cell Therapy for Multiple Sclerosis: Entering an era. Ann Neurol 2018; 83:13-26. [PMID: 29244240 PMCID: PMC5876115 DOI: 10.1002/ana.25119] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 12/01/2017] [Accepted: 12/09/2017] [Indexed: 12/12/2022]
Abstract
Monoclonal antibodies that target CD20 expressing B cells represent an important new treatment option for patients with multiple sclerosis (MS). B-cell-depleting therapy is highly effective against relapsing forms of the disease and is also the first treatment approach proven to protect against disability worsening in primary progressive MS. Moreover, evolving clinical experience with B-cell therapy, combined with a more sophisticated understanding of humoral immunity in preclinical models and in patients with MS, has led to major progress in deciphering the immune pathogenesis of MS. Here, we review the nuanced roles of B cells in MS autoimmunity, the clinical data supporting use of ocrelizumab and other anti-CD20 therapies in the treatment of MS, as well as safety and practical considerations for prescribing. Last, we summarize remaining unanswered questions regarding the proper role of anti-CD20 therapy in MS, its limitations, and the future landscape of B-cell-based approaches to treatment. Ann Neurol 2018;83:13-26.
Collapse
Affiliation(s)
- Ariele L. Greenfield
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, California
| | - Stephen L. Hauser
- Weill Institute for Neurosciences, Department of Neurology, University of California, San Francisco, San Francisco, California
| |
Collapse
|
6
|
Dock J, Ramirez CM, Hultin L, Hausner MA, Hultin P, Elliott J, Yang OO, Anton PA, Jamieson BD, Effros RB. Distinct aging profiles of CD8+ T cells in blood versus gastrointestinal mucosal compartments. PLoS One 2017; 12:e0182498. [PMID: 28832609 PMCID: PMC5568404 DOI: 10.1371/journal.pone.0182498] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/19/2017] [Indexed: 01/10/2023] Open
Abstract
A hallmark of human immunosenescence is the accumulation of late-differentiated memory CD8+ T cells with features of replicative senescence, such as inability to proliferate, absence of CD28 expression, shortened telomeres, loss of telomerase activity, enhanced activation, and increased secretion of inflammatory cytokines. Importantly, oligoclonal expansions of these cells are associated with increased morbidity and mortality risk in elderly humans. Currently, most information on the adaptive immune system is derived from studies using peripheral blood, which contains approximately only 2% of total body lymphocytes. However, most lymphocytes reside in tissues. It is not clear how representative blood changes are of the total immune status. This is especially relevant with regard to the human gastrointestinal tract (GALT), a major reservoir of total body lymphocytes (approximately 60%) and an anatomical region of high antigenic exposure. To assess how peripheral blood T cells relate to those in other locations, we compare CD8+ T cells from peripheral blood and the GALT, specifically rectosigmoid colon, in young/middle age, healthy donors, focusing on phenotypic and functional alterations previously linked to senescence in peripheral blood. Overall, our results indicate that gut CD8+ T cells show profiles suggestive of greater differentiation and activation than those in peripheral blood. Specifically, compared to blood from the same individual, the gut contains significantly greater proportions of CD8+ T cells that are CD45RA- (memory), CD28-, CD45RA-CD28+ (early memory), CD45RA-CD28- (late memory), CD25-, HLA-DR+CD38+ (activated) and Ki-67+ (proliferating); ex vivo CD3+ telomerase activity levels are greater in the gut as well. However, gut CD8+ T cells may not necessarily be more senescent, since they expressed significantly lower levels of CD57 and PD-1 on CD45RO+ memory cells, and had in vitro proliferative dynamics similar to that of blood cells. Compartment-specific age-effects in this cohort were evident as well. Blood cells showed a significant increase with age in proportion of HLA-DR+38+, Ki-67+ and CD25+ CD8+ T cells; and an increase in total CD3+ex-vivo telomerase activity that approached significance. By contrast, the only age-effect seen in the gut was a significant increase in CD45RA- (memory) and concurrent decrease in CD45RA+CD28+ (naïve) CD8+ T cells. Overall, these results indicate dynamics of peripheral blood immune senescence may not hold true in the gut mucosa, underscoring the importance for further study of this immunologically important tissue in evaluating the human immune system, especially in the context of chronic disease and aging.
Collapse
Affiliation(s)
- Jeffrey Dock
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, United States of America
| | - Christina M Ramirez
- Department of Biostatistics, Fielding School of Public Health, University of California-Los Angeles, Los Angeles, CA, United States of America
| | - Lance Hultin
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, United States of America.,UCLA AIDS Institute, David Geffen School of Medicine at UCLA, United States of America
| | - Mary Ann Hausner
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, United States of America.,UCLA AIDS Institute, David Geffen School of Medicine at UCLA, United States of America
| | - Patricia Hultin
- UCLA AIDS Institute, David Geffen School of Medicine at UCLA, United States of America.,Department of Epidemiology, Fielding School of Public Health, University of California-Los Angeles, Los Angeles, CA, United States of America
| | - Julie Elliott
- Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, United States of America.,UCLA AIDS Institute, David Geffen School of Medicine at UCLA, United States of America
| | - Otto O Yang
- UCLA AIDS Institute, David Geffen School of Medicine at UCLA, United States of America.,Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, United States of America.,Department of Microbiology Immunology & Molecular Genetics, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, United States of America.,AIDS Healthcare Foundation, Los Angeles, CA, United States of America
| | - Peter A Anton
- UCLA AIDS Institute, David Geffen School of Medicine at UCLA, United States of America.,Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, United States of America
| | - Beth D Jamieson
- Division of Hematology and Oncology, Department of Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, United States of America.,UCLA AIDS Institute, David Geffen School of Medicine at UCLA, United States of America
| | - Rita B Effros
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, United States of America.,UCLA AIDS Institute, David Geffen School of Medicine at UCLA, United States of America
| |
Collapse
|