Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
Drugs ; 83(16): 1551-1557, 2023 Nov.
Article En | MEDLINE | ID: mdl-37856038

Pozelimab (pozelimab-bbfg; VEOPOZ™) is a fully human immunoglobulin (Ig) G4P (i.e. IgG4 with a proline substitution to promote stabilization of the disulfide bonds between the two heavy chains) monoclonal antibody developed by Regeneron Pharmaceuticals Inc., to block the activity of complement factor 5 (C5) and prevent diseases mediated by the complement pathway. In August 2023, pozelimab received its first approval for the treatment of adults, and paediatric patients aged ≥ 1 year with CD55-deficient protein-losing enteropathy (PLE), also known as CHAPLE disease, in the USA. It is the first US FDA-approved treatment for this disease. In the USA, pozelimab has been granted orphan drug designations for the treatment of paroxysmal nocturnal haemoglobinuria (PNH) [both as a monotherapy and in combination with cemdisiran] and for the treatment of myasthenia gravis (in combination with cemdisiran). Pozelimab is also undergoing clinical development in several other countries worldwide for the treatment of CD55-deficient PLE, PNH and myasthenia gravis. This article summarizes the milestones in the development of pozelimab leading to this first approval for the treatment of adults, and paediatric patients aged ≥ 1 year with CD55-deficient PLE, also known as CHAPLE disease, in the USA.


Hemoglobinuria, Paroxysmal , Myasthenia Gravis , Humans , Child , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal/therapeutic use , Complement System Proteins/therapeutic use , CD55 Antigens/therapeutic use , Complement C5 , Hemoglobinuria, Paroxysmal/drug therapy , Myasthenia Gravis/drug therapy
2.
Blood ; 139(13): 1954-1972, 2022 03 31.
Article En | MEDLINE | ID: mdl-34415298

In 2007 and 2009, the regulatory approval of the first-in-class complement inhibitor eculizumab revolutionized the clinical management of 2 rare, life-threatening clinical conditions: paroxysmal nocturnal hemoglobinuria (PNH) and atypical hemolytic uremic syndrome (aHUS). Although being completely distinct diseases affecting blood cells and the glomerulus, PNH and aHUS remarkably share several features in their etiology and clinical presentation. An imbalance between complement activation and regulation at host surfaces underlies both diseases precipitating in severe thrombotic events that are largely resistant to anticoagulant and/or antiplatelet therapies. Inhibition of the common terminal complement pathway by eculizumab prevents the frequently occurring thrombotic events responsible for the high mortality and morbidity observed in patients not treated with anticomplement therapy. Although many in vitro and ex vivo studies elaborate numerous different molecular interactions between complement activation products and hemostasis, this review focuses on the clinical evidence that links these 2 fields in humans. Several noninfectious conditions with known complement involvement are scrutinized for common patterns concerning a prothrombotic statues and the occurrence of certain complement activation levels. Next to PNH and aHUS, germline-encoded CD59 or CD55 deficiency (the latter causing the disease complement hyperactivation, angiopathic thrombosis, and protein-losing enteropathy), autoimmune hemolytic anemia, (catastrophic) antiphospholipid syndrome, and C3 glomerulopathy are considered. Parallels and distinct features among these conditions are discussed against the background of thrombosis, complement activation, and potential complement diagnostic and therapeutic avenues.


Atypical Hemolytic Uremic Syndrome , Hemoglobinuria, Paroxysmal , Atypical Hemolytic Uremic Syndrome/drug therapy , CD55 Antigens/therapeutic use , Complement Activation , Complement System Proteins/metabolism , Humans
3.
J Immunol ; 198(12): 4837-4845, 2017 06 15.
Article En | MEDLINE | ID: mdl-28500075

Deficiency in the membrane-bound complement regulators CD55 and CD59 exacerbates renal ischemia-reperfusion injury (IRI) in mouse models, but the effect of increasing CD55 and CD59 activity has not been examined. In this study, we investigated the impact of overexpression of human (h) CD55 ± hCD59 or treatment with soluble rhCD55 in a mouse model of renal IRI. Unilaterally nephrectomised mice were subjected to 18 (mild IRI) or 22 min (moderate IRI) warm renal ischemia, and analyzed 24 h after reperfusion for renal function (serum creatinine and urea), complement deposition (C3b/c and C9), and infiltration of neutrophils and macrophages. Transgenic mice expressing hCD55 alone were protected against mild renal IRI, with reduced creatinine and urea levels compared with wild type littermates. However, the renal function of the hCD55 mice was not preserved in the moderate IRI model, despite a reduction in C3b/c and C9 deposition and innate cell infiltration. Mice expressing both hCD55 and hCD59, on the other hand, were protected in the moderate IRI model, with significant reductions in all parameters measured. Wild type mice treated with rhCD55 immediately after reperfusion were also protected in the moderate IRI model. Thus, manipulation of CD55 activity to increase inhibition of the C3 and C5 convertases is protective against renal IRI, and the additional expression of hCD59, which regulates the terminal complement pathway, provides further protection. Therefore, anti-complement therapy using complement regulatory proteins may provide a potential clinical option for preventing tissue and organ damage in renal IRI.


CD55 Antigens/genetics , CD55 Antigens/therapeutic use , CD59 Antigens/genetics , Kidney Diseases/therapy , Reperfusion Injury/therapy , Animals , CD55 Antigens/immunology , Complement Activation , Creatinine/blood , Humans , Kidney Diseases/immunology , Kidney Diseases/physiopathology , Macrophages/immunology , Mice , Mice, Transgenic , Neutrophils/immunology , Reperfusion Injury/immunology , Reperfusion Injury/physiopathology , Urea/blood
4.
Genet Mol Res ; 14(3): 7782-92, 2015 Jul 14.
Article En | MEDLINE | ID: mdl-26214459

It is expensive to induce experimental autoimmune myasthenia gravis (EAMG) by active immunity, and difficult to obtain natural acetylcholine receptor (AChR). We sought a new method of inducing EAMG by immunizing rats with artificially synthesized AChR. The AChR mRNA in TE671 cells was extracted and reverse transcribed. The inclusion body was purified and protein concentration was determined, and the EAMG animal model was used for induction. The serum was extracted from rat blood. The antibody titer was determined using enzyme-linked immunosorbant assay (ELISA). The concentration of decay accelerating factor (DAF) in the rat serum was determined by ELISA, and the metabolism of serum rDAF was determined by western blot. We evaluated the inhibition of rDAF by determining the 50% complement hemolysis unit in the rat serum. The extracellular domain (ECD) nucleotide sequence clone produced by polymerase chain reaction was completely consistent with that in the human gene bank; it was induced by isopropyl ß-D-1-thiogalactopyranoside to express the protein after insertion into vector pET16b. Sodium dodecyl sulfate polyacrylamide gel electrophoresis demonstrated that the inclusion body protein was the exact target. The ECD protein was able to bind with mAb35 after dialysis and renaturation, which demonstrated protein activity. The soluble ECD protein was used to immunize rats and obtain the EAMG models. The inhibitory effect of the complement was unsatisfactory owing to high decay rate after rDAF injection into the EAMG models. It is easy to induce the EAMG model by obtaining the AChRTEα1 subunit ECD protein using the substitution method.


CD55 Antigens/therapeutic use , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Neuroprotective Agents/therapeutic use , Animals , CD55 Antigens/administration & dosage , CD55 Antigens/blood , Disease Models, Animal , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Female , Humans , Injections, Intravenous , Myasthenia Gravis, Autoimmune, Experimental/blood , Myasthenia Gravis, Autoimmune, Experimental/pathology , Neuroprotective Agents/administration & dosage , Neuroprotective Agents/pharmacology , Protein Renaturation/drug effects , Protein Structure, Tertiary , Rats, Inbred Lew , Recombinant Proteins/metabolism , Solubility
5.
J Trauma ; 71(1 Suppl): S151-60, 2011 Jul.
Article En | MEDLINE | ID: mdl-21795874

BACKGROUND: Activation of complement system has been associated with tissue injury after hemorrhage and resuscitation in rats and swine. This study investigated whether administration of human recombinant decay-accelerating factor (DAF; a complement regulatory protein that inhibits classical and alternative pathways) reduces tissue damage in a porcine model of hemorrhagic shock. METHODS: Male Yorkshire swine assigned to four groups were subjected to controlled, isobaric hemorrhage over 15 minutes to a target mean arterial pressure of 35 mm Hg. Hypotension was maintained for 20 minutes followed by a bolus intravenous injection of DAF or vehicle and then animals were observed for 200 minutes. Blood chemistry and physiologic parameters were recorded. Tissue samples from lung and small intestine were subjected to histopathological evaluation and detection of tissue deposition of complement proteins by immunohistochemistry and Western blot analyses. RESULTS: Administration of DAF significantly reduced intestinal and lung tissue damage in a dose-dependent manner (5, 25, and 50 µg/kg). In addition, DAF treatment improved hemorrhage-induced hyperkalemia. The protective effects of DAF appear to be related to its ability to reduce tissue complement activation and deposition on affected tissues. CONCLUSIONS: DAF treatment decreased tissue complement activation and deposition in hemorrhaged animals and attenuated tissue damage at 200 minutes after treatment. The observed beneficial effects of DAF treatment on tissue injury after 20 minutes of severe hypotension presents an attractive model of small volume resuscitation, particularly in situations with a restrictive medical logistical footprint such as far-forward access to first responders in the battlefield or in remote rural or mountainous environments.


CD55 Antigens/therapeutic use , Hyperkalemia/prevention & control , Intestines/pathology , Lung/pathology , Shock, Hemorrhagic/drug therapy , Animals , Blotting, Western , Complement Activation/drug effects , Hemodynamics , Humans , Hyperkalemia/etiology , Intestines/drug effects , Lung/drug effects , Male , Recombinant Proteins/therapeutic use , Resuscitation/methods , Shock, Hemorrhagic/complications , Shock, Hemorrhagic/pathology , Swine , Time Factors
6.
J Surg Res ; 167(2): e103-15, 2011 May 15.
Article En | MEDLINE | ID: mdl-20189588

BACKGROUND: C-reactive protein (CRP) is an acute pro-inflammatory mediator that has been demonstrated to enhance ischemia/reperfusion (IR) injury by virtue of activating the complement system. CRP is able to interact with complement proteins such as C1q, complement factor H, and C4b-binding protein. Since complement activation is central in the expression of tissue injury following IR, we have investigated the effects of human decay-accelerating factor (DAF), a complement inhibitor, on CRP-potentiated complement activation and tissue injury in mice subjected to mesenteric IR. MATERIALS AND METHODS: Male C57B1/6 mice were allocated into eight groups: (1) Sham-operated group without IR injury; (2) CRP+Sham group; (3) IR group; (4) CRP+IR group; (5) DAF group; (6) CRP+DAF group; (7) IR+DAF group, and (8) CRP+IR+DAF group. Intestinal and lung injury, neutrophil infiltration, myeloperoxidase (MPO) expression, complement component deposition, and interleukin-6 (IL-6) production were assessed for each treatment group of mice. RESULTS: We report that administration of DAF significantly attenuates the CRP-enhanced intestinal injury as well as remote lung damages following acute mesenteric IR in mice, while DAF inhibits complement activation, suppresses neutrophil infiltration, and reduces IL-6 production. CONCLUSIONS: Our study suggests that inhibition complement activation with DAF may prove useful for the treatment of post-ischemic inflammatory injuries associated with an increased production of CRP.


C-Reactive Protein/metabolism , CD55 Antigens/therapeutic use , Mesentery/metabolism , Mesentery/physiopathology , Reperfusion Injury/metabolism , Reperfusion Injury/prevention & control , Animals , CD55 Antigens/administration & dosage , CD55 Antigens/pharmacology , Complement C5a/metabolism , Complement Membrane Attack Complex/metabolism , Injections, Intravenous , Interleukin-6/metabolism , Male , Mesentery/blood supply , Mice , Mice, Inbred C57BL , Models, Animal , Neutrophil Infiltration/drug effects , Peroxidase/metabolism , Reperfusion Injury/physiopathology
7.
Invest Ophthalmol Vis Sci ; 51(12): 6776-83, 2010 Dec.
Article En | MEDLINE | ID: mdl-20688727

PURPOSE: Sequence variations in complement proteins are associated with age-related macular degeneration (AMD). The terminal pathway of complement results in the formation of the membrane attack complex (MAC) on the cell surface, resulting in their lysis. MAC has been documented on the retinal pigment epithelium (RPE), choroidal blood vessels, and drusen of AMD eyes. Here the investigators test the hypothesis that increasing the expression of decay accelerating factor (CD55) on RPE cells may result in reduced MAC-mediated damage. METHODS: The investigators constructed a recombinant adenovirus expressing human CD55 (AdCAGCD55). Mouse hepatocytes were infected with AdCAGCD55 or negative controls and subsequently incubated with normal human serum (NHS). Cell lysis and MAC formation were measured by FACS and immunocytochemistry, respectively. Adult mice were injected in the subretinal space with either AdCAGCD55 or controls; after 1 week of CD55 transgene expression, the eyecups were excised, challenged with NHS, and quantified for human MAC formation. RESULTS: Control-infected or uninfected mouse hepatocytes lyse at a rate of 93% and 94%, respectively. AdCAGCD55- infected mouse hepatocytes lyse at a rate of 29%. Lysis was confirmed to occur in the presence of MAC, which was reduced by 67% when cells were infected by AdCAGCD55. Mice injected in the subretinal space with AdCAGCD55 exhibited a 55.7% reduction in MAC formation on the RPE relative to controls. CONCLUSIONS: Adenovirus-mediated delivery of hCD55 to murine RPE confers protection against human complement. The investigators propose that the expression of hCD55 on RPE cells warrants investigation as a potential therapy for AMD.


CD55 Antigens/therapeutic use , Complement Membrane Attack Complex/metabolism , Complement System Proteins/physiology , Genetic Therapy , Macular Degeneration/therapy , Retinal Pigment Epithelium/metabolism , Adenoviridae/genetics , Animals , Blotting, Western , CD55 Antigens/genetics , Cell Line , Flow Cytometry , Gene Expression/physiology , Genetic Vectors , Hemolysis/physiology , Hepatocytes/metabolism , Hepatocytes/virology , Humans , Immunohistochemistry , Macular Degeneration/metabolism , Mice , Mice, Inbred C57BL , Transgenes
8.
Protein Expr Purif ; 66(1): 102-6, 2009 Jul.
Article En | MEDLINE | ID: mdl-19254764

Decay accelerating factor (DAF) is a very potent complement regulatory protein which holds promise for clinical usage. Here we report on an improved procedure for refolding both rat and human DAF over-expressed in Escherichia coli. It was shown that 50-70% of the inclusion body could be refolded to soluble active protein. This method excludes the use of L-arginine, which is expensive, and can be used to prepare a large quantity of recombinant DAF for therapeutic studies.


CD55 Antigens/chemistry , CD55 Antigens/metabolism , Protein Folding , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Animals , Arginine/chemistry , CD55 Antigens/genetics , CD55 Antigens/therapeutic use , Cloning, Molecular , HeLa Cells , Humans , Inclusion Bodies/chemistry , Inclusion Bodies/metabolism , Protein Renaturation , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/therapeutic use
9.
J Immunol ; 177(8): 5558-66, 2006 Oct 15.
Article En | MEDLINE | ID: mdl-17015743

Decay-accelerating factor (DAF) and complement receptor 1-related gene/protein y (Crry) are two membrane-bound complement regulators on murine erythrocytes that inhibit C3/C5 convertases. Previously, we found that Crry- but not DAF-deficient erythrocytes were susceptible to alternative pathway complement-mediated elimination in vivo. To determine whether it is a unique activity or a higher level expression of Crry makes it indispensable on murine erythrocytes, we over-expressed DAF on Crry-deficient (Crry(-/-)) erythrocytes by retroviral vector-mediated DAF gene transduction of bone marrow stem cells. DAF retrovirus-transduced erythrocytes expressed 846 +/- 127 DAF molecules/cell (DAF(high)) compared with 249 +/- 94 DAF molecules/cell (DAF(low)) and 774 +/- 135 Crry molecules/cell on control mouse erythrocytes. DAF(high)-Crry(-/-) erythrocytes were significantly more resistant than either DAF(low)-Crry(-/-), DAF(-/-) -Crry(+/+) or wild-type erythrocytes to classical pathway complement-mediated C3 deposition in vitro. Furthermore, increased DAF expression rescued Crry(-/-) erythrocytes from acute alternative pathway complement attack in vivo. Notably, long term monitoring revealed that DAF(high)-Crry(-/-) erythrocytes were still more susceptible than wild-type erythrocytes to complement-mediated elimination as they had a shorter half-life in complement-sufficient mice but survived equally well in complement-deficient mice. These results suggest that both a high level expression and a more potent anti-alternative pathway complement activity of Crry contributed to its indispensable role on murine erythrocytes. Additionally, they demonstrate the feasibility of using stem cell gene therapy to correct membrane complement regulator deficiency on blood cells in vivo.


CD55 Antigens/administration & dosage , Complement Pathway, Alternative , Erythrocytes , Receptors, Complement/deficiency , Stem Cells/metabolism , Transduction, Genetic/methods , Animals , Bone Marrow Cells , CD55 Antigens/therapeutic use , Erythrocytes/immunology , Erythrocytes/metabolism , Erythrocytes/pathology , Genetic Therapy/methods , Half-Life , Mice , Mice, Knockout , Receptors, Complement/immunology , Receptors, Complement 3b , Retroviridae
10.
Lab Invest ; 83(1): 75-85, 2003 Jan.
Article En | MEDLINE | ID: mdl-12533688

Coxsackievirus B3 (CVB3) infection can result in myocarditis, which in turn may lead to a protracted immune response and subsequent dilated cardiomyopathy. Human decay-accelerating factor (DAF), a binding receptor for CVB3, was synthesized as a soluble IgG1-Fc fusion protein (DAF-Fc). In vitro, DAF-Fc was able to inhibit complement activity and block infection by CVB3, although blockade of infection varied widely among strains of CVB3. To determine the effects of DAF-Fc in vivo, 40 adolescent A/J mice were infected with a myopathic strain of CVB3 and given DAF-Fc treatment 3 days before infection, during infection, or 3 days after infection; the mice were compared with virus alone and sham-infected animals. Sections of heart, spleen, kidney, pancreas, and liver were stained with hematoxylin and eosin and submitted to in situ hybridization for both positive-strand and negative-strand viral RNA to determine the extent of myocarditis and viral infection, respectively. Salient histopathologic features, including myocardial lesion area, cell death, calcification and inflammatory cell infiltration, pancreatitis, and hepatitis were scored without knowledge of the experimental groups. DAF-Fc treatment of mice either preceding or concurrent with CVB3 infection resulted in a significant decrease in myocardial lesion area and cell death and a reduction in the presence of viral RNA. All DAF-Fc treatment groups had reduced infectious CVB3 recoverable from the heart after infection. DAF-Fc may be a novel therapeutic agent for active myocarditis and acute dilated cardiomyopathy if given early in the infectious period, although more studies are needed to determine its mechanism and efficacy.


CD55 Antigens/therapeutic use , Cardiomyopathy, Dilated/prevention & control , Enterovirus B, Human/isolation & purification , Myocardium/pathology , Viral Load , Animals , Cardiomyopathy, Dilated/virology , Complement System Proteins/physiology , Enterovirus B, Human/genetics , Enterovirus B, Human/growth & development , Mice , RNA, Viral/analysis , Recombinant Proteins/therapeutic use , Viral Plaque Assay
11.
Clin Exp Immunol ; 129(2): 198-207, 2002 Aug.
Article En | MEDLINE | ID: mdl-12165074

Complement activation and subsequent generation of inflammatory molecules and membrane attack complex contributes to the pathology of a number of inflammatory and degenerative diseases, including arthritis, glomerulonephritis and demyelination. Agents that specifically inhibit complement activation might prove beneficial in the treatment of these diseases. Soluble recombinant forms of the naturally occurring membrane complement regulatory proteins (CRP) have been exploited for this purpose. We have undertaken to design better therapeutics based on CRP. Here we describe the generation of soluble, recombinant CRP comprising rat decay accelerating factor (DAF) or rat CD59 expressed as Fc fusion proteins, antibody-like molecules comprising two CRP moieties in place of the antibody Fab arms (CRP-Ig). Reagents bearing DAF on each arm (DAF-Ig), CD59 on each arm (CD59-Ig) and a hybrid reagent containing both DAF and CD59 were generated. All three reagents inhibited C activation in vitro. Compared with soluble CRP lacking Fc domains, activity was reduced, but was fully restored by enzymatic release of the regulator from the Ig moiety, implicating steric constraints in reducing functional activity. In vivo studies showed that DAF-Ig, when compared to soluble DAF, had a much extended half-life in the circulation in rats and concomitantly caused a sustained reduction in plasma complement activity. When given intra-articularly to rats in a model of arthritis, DAF-Ig significantly reduced severity of disease. The data demonstrate the potential of CRP-Ig as reagents for sustained therapy of inflammatory disorders, including arthritis, but emphasize the need for careful design of fusion proteins to retain function.


Complement Inactivator Proteins/metabolism , Immunoglobulins/metabolism , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , CD55 Antigens/genetics , CD55 Antigens/metabolism , CD55 Antigens/therapeutic use , CHO Cells , Complement Inactivator Proteins/genetics , Complement Inactivator Proteins/therapeutic use , Cricetinae , Half-Life , Hemolysis , Humans , Immunoglobulins/genetics , Immunoglobulins/therapeutic use , In Vitro Techniques , Indicators and Reagents , Rats , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use
12.
Blood ; 92(11): 4439-45, 1998 Dec 01.
Article En | MEDLINE | ID: mdl-9834251

Hemolytic anemia is a major feature of paroxysmal nocturnal hemoglobinuria (PNH). Intravascular red blood cell (RBC) destruction is caused by increased sensitivity of the abnormal erythrocyte to complement-mediated lysis, due to the GPI absence of a membrane-bound glycosylphosphatidylinositol (GPI)-linked protein, which functions as an inhibitor of reactive lysis (CD59). Both in vivo and in vitro models have suggested the feasibility of cell-to-cell transfer of GPI proteins, and patients with hemolysis could potentially benefit from transfer of CD59 to their deficient erythrocytes. We studied the ability of RBC components prepared from outdated packed RBC collections, as well as high-density lipoprotein (HDL) preparations, rich in CD55 and CD59, to promote protein transfer, as assessed by flow cytometry, immunoblotting, and susceptibility to complement-mediated lysis. By flow cytometry, CD55 and CD59 were present on RBC-derived microvesicles that stained with an antiglycophorin antibody Ab; in addition, soluble CD59 and CD55 were detected by immunoblot in soluble fractions eluated from RBC units stored for more than 35 days, but not in fresh blood. Both commercial HDL preparations and those prepared in our laboratory contained CD55 and CD59, as assayed by immunoblot. When RBC that were deficient (GPI)-anchored protein, obtained from five patients, with PNH were incubated with HDL preparations for 2 to 4 hours, there was significant transfer of both proteins to the cell surface, as demonstrated by flow cytometry. Washed RBC microvesicles, prepared by ultrasonification, also mediated transfer of GPI-linked proteins to deficient RBC. Pretreatment of microvesicles, RBC eluate preparations, and HDL with phosphatidylinositol-specific, phospholipase C, abrogated protein transfer to deficient cells, indicating that increased cell-associated CD55 and CD59 levels were related to insertion of the intact GPI moiety, rather than to simple adhesion. PNH RBC that were exposed to HDL, RBC eluate preparations, or microvesicles demonstrated decreased in vitro complement-mediated hemolysis in the Ham test. Transfer of GPI-linked proteins from soluble preparations containing CD55 and CD59 to PNH erythrocytes is feasible and may have clinical utility.


CD55 Antigens/administration & dosage , CD59 Antigens/administration & dosage , Drug Delivery Systems , Erythrocytes/metabolism , Hemoglobinuria, Paroxysmal/blood , CD55 Antigens/immunology , CD55 Antigens/metabolism , CD55 Antigens/therapeutic use , CD59 Antigens/immunology , CD59 Antigens/metabolism , CD59 Antigens/therapeutic use , Erythrocytes/immunology , Erythrocytes/pathology , Flow Cytometry , Glycosylphosphatidylinositols , Hemoglobinuria, Paroxysmal/drug therapy , Humans , Lipoproteins, HDL
...