1
|
Taha S, Fantoni G, Hong E, Terrade A, Doucoure O, Deghmane AE, Taha MK. Characterization of Unusual Serogroups of Neisseria meningitidis. Microorganisms 2024; 12:2528. [PMID: 39770731 PMCID: PMC11676732 DOI: 10.3390/microorganisms12122528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Most cases of invasive meningococcal disease (IMD) in Europe are caused by isolates of the Neisseria meningitidis serogroups B, C, W, and Y. We aimed to explore cases caused by other unusual serogroups. We retrospectively screened IMD cases in the databases of the National Reference Center for Meningococci and Haemophilus influnezae in France between 2014 and 2023. Age, sex, serogroups, and genetic lineage distributions were analyzed. We also measured complement deposition on the bacterial surface and tested coverage by vaccines against serogroup B. Cases due to isolates of serogroups other than B, C, W, and Y represented 1.6% of all 3610 IMD cases during the study period with 59 cases and a median age of 21.5 years of age. The corresponding isolates were non-groupable (26 cases), serogroup X (21 cases), serogroup E (11 cases), and one isolate belonged to serogroup Z. Only a low proportion (7.4%) belonged to the hyperinvasive genetic lineages. Isolates of serogroup E bound a significantly higher amount of complement on their surface and were mainly detected in patients with terminal complement pathway deficiencies. Isolates of these unusual serogroups were shown to be covered by vaccines licensed against meningococci B. Surveillance of these isolates needs to be enhanced.
Collapse
Affiliation(s)
- Samy Taha
- Institut Pasteur, Invasive Bacterial Infections, Université Paris Cité, 75015 Paris, France; (G.F.); (E.H.); (A.T.); (O.D.); (A.-E.D.)
| | - Giulia Fantoni
- Institut Pasteur, Invasive Bacterial Infections, Université Paris Cité, 75015 Paris, France; (G.F.); (E.H.); (A.T.); (O.D.); (A.-E.D.)
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Eva Hong
- Institut Pasteur, Invasive Bacterial Infections, Université Paris Cité, 75015 Paris, France; (G.F.); (E.H.); (A.T.); (O.D.); (A.-E.D.)
| | - Aude Terrade
- Institut Pasteur, Invasive Bacterial Infections, Université Paris Cité, 75015 Paris, France; (G.F.); (E.H.); (A.T.); (O.D.); (A.-E.D.)
| | - Oumar Doucoure
- Institut Pasteur, Invasive Bacterial Infections, Université Paris Cité, 75015 Paris, France; (G.F.); (E.H.); (A.T.); (O.D.); (A.-E.D.)
| | - Ala-Eddine Deghmane
- Institut Pasteur, Invasive Bacterial Infections, Université Paris Cité, 75015 Paris, France; (G.F.); (E.H.); (A.T.); (O.D.); (A.-E.D.)
| | - Muhamed-Kheir Taha
- Institut Pasteur, Invasive Bacterial Infections, Université Paris Cité, 75015 Paris, France; (G.F.); (E.H.); (A.T.); (O.D.); (A.-E.D.)
| |
Collapse
|
2
|
Gonul S, Eker S. Bilateral Retinal Venous Occlusion in Atypical Hemolytic-Uremic Syndrome Due to Complement Factor H Mutation. Ocul Immunol Inflamm 2024; 32:2285-2289. [PMID: 38536978 DOI: 10.1080/09273948.2024.2333401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/15/2024] [Accepted: 03/17/2024] [Indexed: 08/13/2024]
Abstract
PURPOSE Atypical hemolytic uremic syndrome (aHUS) is a rare progressive thrombotic microangiopathy caused by overactivation in the alternative complement pathway. A wide spectrum of environmental triggers, such as viruses, vaccination, drugs, pregnancy, neoplasms, transplant, and autoimmune diseases can cause aHUS in genetically susceptible individuals. In this report, the diagnosis and treatment process of aHUS and bilateral retinal venous occlusion (RVO) will be presented. METHODS Single-case, retrospective management of ophthalmological and systemic manifestations. RESULTS A 28-year-old G2P2 female with acute blurred vision and history of acute renal failure. She was diagnosed with preeclampsia in her gestation history. After the laboratory work-up, the diagnosis of aHUS was confirmed. She was treated with eculizumab following 14 days of plasmapheresis. However, her visual acuity was 20/20 on the right and 20/60 on the left at the time of admission. Retinal examination revealed flame-shaped hemorrhages, exudation, and macular edema. The patient was diagnosed with branch RVO in the right eye. Subsequently, central RVO was occurred in the left eye. Intravitreal dexamethasone implant was administered for both eyes since there was no reasonable regression in retinal findings with bevacizumab treatment. She went into remission and her BCVA reached 20/25 during the 12-month follow-up period under the eculizumab therapy. CONCLUSION Diagnosis of aHUS is challenging especially during pregnancy and the postpartum period. Although ocular involvement is quite rare, we described bilateral RVO in aHUS case with homozygous nonsense mutation (c.2134 G > T p.G712). Dexamethasone implant should be considered for the treatment of RVO in aHUS cases.
Collapse
Affiliation(s)
- Saban Gonul
- Department of Ophthalmology, Selcuk University Faculty of Medicine, Konya, Turkey
| | - Serhat Eker
- Department of Ophthalmology, Isparta Yalvaç State Hospital, Isparta, Turkey
| |
Collapse
|
3
|
Qi J, Wu T, Wang J, Zhang J, Chen L, Jiang Z, Li Y, Jiang H, Sun Q, Gu Q, Ying Z. Research trends and frontiers in lupus nephritis: a bibliometric analysis from 2012 to 2022. Int Urol Nephrol 2024; 56:781-794. [PMID: 37581688 PMCID: PMC10808147 DOI: 10.1007/s11255-023-03715-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/16/2023] [Indexed: 08/16/2023]
Abstract
OBJECTIVES Lupus nephritis is a prevalent renal manifestation of systemic lupus erythematosus (SLE) and represents a significant cause of morbidity and mortality associated with the disease. This study endeavors to undertake a meticulous bibliometric analysis of LN publications to comprehend the research hotspots and future directions. METHODS The literature on LN was acquired from the Web of Science Core Collection (WoSCC). Co-occurrence and cooperative relationship analysis of authors, institutions, countries, journals, references and keywords in the publication was performed through CiteSpace, VOSviewer and a bibliometric online analysis platform. The knowledge graphs were created, and clustering and emergence analyses were performed. RESULTS According to the search strategy, a total of 2077 publications related to lupus nephritis (LN) have been identified, with China being the largest contributor globally. The Ohio State University emerged as the most prolific institution. Lupus is the most cited and published journal. Jan J Weening and Brad Rovin were the most prolific and cocited authors. The current research focus revolved around the "nirp3 inflammasome," "biomarker," and "voclosporin". "international society," "thrombotic microangiopathy (TMA)," and "pathway" were identified to be future research hotpots by keyword burst analysis. CONCLUSIONS This bibliometric analysis summarizes for the first time the progress of LN research (2012-2022), and qualitatively and quantitatively evaluates the bibliometric information of LN research. There has been a steady increase in the scientific literature on LN over the past 11 years, with an average growth rate of 7.27%. In this field, researchers are primarily based in China and the United States. The pathogenic mechanisms, management strategies and prognostic outcomes of LN are acknowledged as prospective research hotspots. Bibliometrically, the research status and trends of LN publications may greatly assist and be a significant reference for future research in the area.
Collapse
Affiliation(s)
- Jiaping Qi
- Center for General Practice Medicine, Department of Rheumatology and Immunology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158, Shangtang Road, Hangzhou, 310014, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Cultivation for Arthritis Diagnosis and Treatment, Hangzhou, 310000, Zhejiang, China
- Bengbu Medical College, Bengbu, China
| | - Teng Wu
- Department of Rheumatology, Affiliated Hospital of Nantong University, Nantong, China
- Dalian Medical University, Dalian, China
| | - Jing Wang
- Center for General Practice Medicine, Department of Rheumatology and Immunology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158, Shangtang Road, Hangzhou, 310014, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Cultivation for Arthritis Diagnosis and Treatment, Hangzhou, 310000, Zhejiang, China
| | - Ju Zhang
- Center for General Practice Medicine, Department of Rheumatology and Immunology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158, Shangtang Road, Hangzhou, 310014, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Cultivation for Arthritis Diagnosis and Treatment, Hangzhou, 310000, Zhejiang, China
| | - Lin Chen
- Center for General Practice Medicine, Department of Rheumatology and Immunology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158, Shangtang Road, Hangzhou, 310014, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Cultivation for Arthritis Diagnosis and Treatment, Hangzhou, 310000, Zhejiang, China
| | - Zhaoyu Jiang
- Center for General Practice Medicine, Department of Rheumatology and Immunology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158, Shangtang Road, Hangzhou, 310014, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Cultivation for Arthritis Diagnosis and Treatment, Hangzhou, 310000, Zhejiang, China
| | - Yixuan Li
- Center for General Practice Medicine, Department of Rheumatology and Immunology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158, Shangtang Road, Hangzhou, 310014, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Cultivation for Arthritis Diagnosis and Treatment, Hangzhou, 310000, Zhejiang, China
| | - Huan Jiang
- Center for General Practice Medicine, Department of Rheumatology and Immunology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158, Shangtang Road, Hangzhou, 310014, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Cultivation for Arthritis Diagnosis and Treatment, Hangzhou, 310000, Zhejiang, China
| | - Qiong Sun
- Center for General Practice Medicine, Department of Rheumatology and Immunology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158, Shangtang Road, Hangzhou, 310014, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Cultivation for Arthritis Diagnosis and Treatment, Hangzhou, 310000, Zhejiang, China
| | - Qingchen Gu
- Center for General Practice Medicine, Department of Rheumatology and Immunology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158, Shangtang Road, Hangzhou, 310014, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Cultivation for Arthritis Diagnosis and Treatment, Hangzhou, 310000, Zhejiang, China
| | - Zhenhua Ying
- Center for General Practice Medicine, Department of Rheumatology and Immunology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158, Shangtang Road, Hangzhou, 310014, Zhejiang, China.
- Zhejiang Provincial Key Laboratory of Traditional Chinese Medicine Cultivation for Arthritis Diagnosis and Treatment, Hangzhou, 310000, Zhejiang, China.
- Bengbu Medical College, Bengbu, China.
| |
Collapse
|
4
|
Kolev M, Barbour T, Baver S, Francois C, Deschatelets P. With complements: C3 inhibition in the clinic. Immunol Rev 2023; 313:358-375. [PMID: 36161656 DOI: 10.1111/imr.13138] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
C3 is a key complement protein, located at the nexus of all complement activation pathways. Extracellular, tissue, cell-derived, and intracellular C3 plays critical roles in the immune response that is dysregulated in many diseases, making it an attractive therapeutic target. However, challenges such as very high concentration in blood, increased acute expression, and the elevated risk of infections have historically posed significant challenges in the development of C3-targeted therapeutics. This is further complicated because C3 activation fragments and their receptors trigger a complex network of downstream effects; therefore, a clear understanding of these is needed to provide context for a better understanding of the mechanism of action (MoA) of C3 inhibitors, such as pegcetacoplan. Because of C3's differential upstream position to C5 in the complement cascade, there are mechanistic differences between pegcetacoplan and eculizumab that determine their efficacy in patients with paroxysmal nocturnal hemoglobinuria. In this review, we compare the MoA of pegcetacoplan and eculizumab in paroxysmal nocturnal hemoglobinuria and discuss the complement-mediated disease that might be amenable to C3 inhibition. We further discuss the current state and outlook for C3-targeted therapeutics and provide our perspective on which diseases might be the next success stories in the C3 therapeutics journey.
Collapse
Affiliation(s)
- Martin Kolev
- Apellis Pharmaceuticals, Waltham, Massachusetts, USA
| | - Tara Barbour
- Apellis Pharmaceuticals, Waltham, Massachusetts, USA
| | - Scott Baver
- Apellis Pharmaceuticals, Waltham, Massachusetts, USA
| | | | | |
Collapse
|
5
|
Rysava R, Peiskerova M, Tesar V, Benes J, Kment M, Szilágyi Á, Csuka D, Prohászka Z. Atypical hemolytic uremic syndrome triggered by mRNA vaccination against SARS-CoV-2: Case report. Front Immunol 2022; 13:1001366. [PMID: 36275662 PMCID: PMC9580272 DOI: 10.3389/fimmu.2022.1001366] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/05/2022] [Indexed: 11/21/2022] Open
Abstract
Atypical hemolytic uremic syndrome (aHUS), also called complement-mediated hemolytic uremic syndrome (CM-HUS), is a rare disease caused by dysregulation in the alternative complement activation pathway. It is a life-threatening condition causing ischemia of a number of organs, and it typically causes acute kidney injury. This disorder may be triggered by various factors including viral or bacterial infections, pregnancy, surgery, and injuries. In about 60% of cases, the genetic origin of the disease can be identified—commonly mutations affecting complementary factor H and MCP protein. Eculizumab, a monoclonal antibody to the C5 component of the complement, represents the current effective treatment.We describe a case of a young woman with a previous history of polyvalent allergies, who developed atypical hemolytic uremic syndrome after vaccination with mRNA vaccine against SARS-CoV-2. The disease manifested by scleral bleeding, acute renal insufficiency, anemia, and thrombocytopenia. The patient was treated with plasma exchanges without sufficient effect; remission occurred only after starting treatment with eculizumab. Genetic examination showed that the patient is a carrier of multiple inherited risk factors (a rare pathogenic variant in CFH, MCPggaac haplotype of the CD46 gene, and the risk haplotype CFH H3). The patient is currently in hematological remission with persistent mild renal insufficiency, continuing treatment with eculizumab/ravulizumab. By this case report, we meant to point out the need for careful monitoring of people after vaccination, as it may trigger immune-mediated diseases, especially in those with predisposing factors.
Collapse
Affiliation(s)
- Romana Rysava
- Department of Nephrology, First Faculty of Medicine, General University Hospital, Charles University, Prague, Czechia
- *Correspondence: Romana Rysava, ; Zoltán Prohászka,
| | - Martina Peiskerova
- Department of Nephrology, First Faculty of Medicine, General University Hospital, Charles University, Prague, Czechia
| | - Vladimir Tesar
- Department of Nephrology, First Faculty of Medicine, General University Hospital, Charles University, Prague, Czechia
| | - Jan Benes
- University Hospital, Charles University – Faculty of Medicine, Hradec Králové, Czechia
- Department of Anesthesiology, Perioperative Medicine and Intensive Care, Masaryk Hospital, Jana Evangelisty (JE) Purkinje University, Ústi nad Labem, Czechia
| | - Martin Kment
- Department of Clinical and Transplant Pathology, Institute of Clinical and Experimental Medicine (IKEM), Prague, Czechia
| | - Ágnes Szilágyi
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest, Hungary
- Research Group for Immunology and Haematology, Eotvos Lorand Research Network (Office for Supported Research Groups), Semmelweis University, Budapest, Hungary
| | - Dorottya Csuka
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest, Hungary
- Research Group for Immunology and Haematology, Eotvos Lorand Research Network (Office for Supported Research Groups), Semmelweis University, Budapest, Hungary
| | - Zoltán Prohászka
- Department of Internal Medicine and Hematology, Semmelweis University, Budapest, Hungary
- Research Group for Immunology and Haematology, Eotvos Lorand Research Network (Office for Supported Research Groups), Semmelweis University, Budapest, Hungary
- *Correspondence: Romana Rysava, ; Zoltán Prohászka,
| |
Collapse
|
6
|
Chai JN, Azad AK, Kuan K, Guo X, Wang Y. A Splice Site Mutation Associated with Congenital CD59 Deficiency. Hematol Rep 2022; 14:172-178. [PMID: 35735736 PMCID: PMC9222317 DOI: 10.3390/hematolrep14020025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 03/23/2022] [Accepted: 05/25/2022] [Indexed: 11/16/2022] Open
Abstract
Congenital CD59 deficiency is a recently described rare autosomal recessive disease associated with CD59 gene mutations that lead to deficient or dysfunctional CD59 protein on the cell surface. The disease is characterized by the early onset of chronic hemolysis, relapsing peripheral demyelinating neuropathy, and recurrent ischemic strokes. To date, there are 14 patients with 4 exon mutations reported globally. A young boy with early onset peripheral neuropathy and atypical hemolytic uremic syndrome is presented. Next-generation sequencing (NGS) identified a homozygous splice site variant in intron 1 of the CD59 gene (c.67 + 1G > T). This variant alters a consensus donor splicing site. Quantitative reverse transcription PCR showed that CD59 mRNA expression in the patient is significantly reduced to 0.017-fold compared to the controls. Flow cytometry showed the lack of CD59 protein on the surface of the patient’s red blood cells. This variant is the first splice site mutation reported to be associated with congenital CD59 deficiency.
Collapse
Affiliation(s)
| | | | | | | | - Yanhua Wang
- Correspondence: ; Tel.: +1-718-920-4976; Fax: +1-718-920-7611
| |
Collapse
|
7
|
Tiller G, Lammerts RGM, Karijosemito JJ, Alkaff FF, Diepstra A, Pol RA, Meter-Arkema AH, Seelen MA, van den Heuvel MC, Hepkema BG, Daha MR, van den Born J, Berger SP. Weak Expression of Terminal Complement in Active Antibody-Mediated Rejection of the Kidney. Front Immunol 2022; 13:845301. [PMID: 35493506 PMCID: PMC9044906 DOI: 10.3389/fimmu.2022.845301] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundThe role of the complement system in antibody-mediated rejection (ABMR) is insufficiently understood. We aimed to investigate the role of local and systemic complement activation in active (aABMR). We quantified complement activation markers, C3, C3d, and C5b-9 in plasma of aABMR, and acute T-cell mediated rejection (aTCMR), and non-rejection kidney transplant recipients. Intra-renal complement markers were analyzed as C4d, C3d, C5b-9, and CD59 deposition. We examined in vitro complement activation and CD59 expression on renal endothelial cells upon incubation with human leukocyte antigen antibodies.MethodsWe included 50 kidney transplant recipients, who we histopathologically classified as aABMR (n=17), aTCMR (n=18), and non-rejection patients (n=15).ResultsComplement activation in plasma did not differ across groups. C3d and C4d deposition were discriminative for aABMR diagnosis. Particularly, C3d deposition was stronger in glomerular (P<0,01), and peritubular capillaries (P<0,05) comparing aABMR to aTCMR rejection and non-rejection biopsies. In contrast to C3d, C5b-9 was only mildly expressed across all groups. For C5b-9, no significant difference between aABMR and non-rejection biopsies regarding peritubular and glomerular C5b-9 deposition was evident. We replicated these findings in vitro using renal endothelial cells and found complement pathway activation with C4d and C3d, but without terminal C5b-9 deposition. Complement regulator CD59 was variably present in biopsies and constitutively expressed on renal endothelial cells in vitro.ConclusionOur results indicate that terminal complement might only play a minor role in late aABMR, possibly indicating the need to re-evaluate the applicability of terminal complement inhibitors as treatment for aABMR.
Collapse
Affiliation(s)
- Gesa Tiller
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, Netherlands
| | - Rosa G. M. Lammerts
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, Netherlands
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Jessy J. Karijosemito
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, Netherlands
| | - Firas F. Alkaff
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, Netherlands
- Division of Pharmacology and Therapy, Department of Anatomy, Histology, and Pharmacology, Faculty of Medicine Universitas Airlangga, Surabaya, Indonesia
| | - Arjan Diepstra
- Division of Pathology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Robert A. Pol
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Anita H. Meter-Arkema
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, Netherlands
| | - Marc. A. Seelen
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, Netherlands
| | - Marius C. van den Heuvel
- Division of Pathology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, Netherlands
| | - Bouke G. Hepkema
- Department of Laboratory Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Mohamed R. Daha
- Department of Nephrology, University of Leiden, Leiden, Netherlands
| | - Jacob van den Born
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, Netherlands
| | - Stefan P. Berger
- Division of Nephrology, Department of Internal Medicine, University Medical Center Groningen, Groningen, Netherlands
- *Correspondence: Stefan P. Berger,
| |
Collapse
|
8
|
Apostolidis SA, Sarkar A, Giannini HM, Goel RR, Mathew D, Suzuki A, Baxter AE, Greenplate AR, Alanio C, Abdel-Hakeem M, Oldridge DA, Giles JR, Wu JE, Chen Z, Huang YJ, Belman J, Pattekar A, Manne S, Kuthuru O, Dougherty J, Weiderhold B, Weisman AR, Ittner CAG, Gouma S, Dunbar D, Frank I, Huang AC, Vella LA, Reilly JP, Hensley SE, Rauova L, Zhao L, Meyer NJ, Poncz M, Abrams CS, Wherry EJ. Signaling Through FcγRIIA and the C5a-C5aR Pathway Mediate Platelet Hyperactivation in COVID-19. Front Immunol 2022; 13:834988. [PMID: 35309299 PMCID: PMC8928747 DOI: 10.3389/fimmu.2022.834988] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/07/2022] [Indexed: 12/12/2022] Open
Abstract
Patients with COVID-19 present with a wide variety of clinical manifestations. Thromboembolic events constitute a significant cause of morbidity and mortality in patients infected with SARS-CoV-2. Severe COVID-19 has been associated with hyperinflammation and pre-existing cardiovascular disease. Platelets are important mediators and sensors of inflammation and are directly affected by cardiovascular stressors. In this report, we found that platelets from severely ill, hospitalized COVID-19 patients exhibited higher basal levels of activation measured by P-selectin surface expression and had poor functional reserve upon in vitro stimulation. To investigate this question in more detail, we developed an assay to assess the capacity of plasma from COVID-19 patients to activate platelets from healthy donors. Platelet activation was a common feature of plasma from COVID-19 patients and correlated with key measures of clinical outcome including kidney and liver injury, and APACHEIII scores. Further, we identified ferritin as a pivotal clinical marker associated with platelet hyperactivation. The COVID-19 plasma-mediated effect on control platelets was highest for patients that subsequently developed inpatient thrombotic events. Proteomic analysis of plasma from COVID-19 patients identified key mediators of inflammation and cardiovascular disease that positively correlated with in vitro platelet activation. Mechanistically, blocking the signaling of the FcγRIIa-Syk and C5a-C5aR pathways on platelets, using antibody-mediated neutralization, IgG depletion or the Syk inhibitor fostamatinib, reversed this hyperactivity driven by COVID-19 plasma and prevented platelet aggregation in endothelial microfluidic chamber conditions. These data identified these potentially actionable pathways as central for platelet activation and/or vascular complications and clinical outcomes in COVID-19 patients. In conclusion, we reveal a key role of platelet-mediated immunothrombosis in COVID-19 and identify distinct, clinically relevant, targetable signaling pathways that mediate this effect.
Collapse
Affiliation(s)
- Sokratis A. Apostolidis
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Division of Rheumatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Amrita Sarkar
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Heather M. Giannini
- Division of Pulmonary, Allergy and Critical Care Medicine, Center for Translational Lung Biology, Lung Biology Institute, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Rishi R. Goel
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Divij Mathew
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Aae Suzuki
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - Amy E. Baxter
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Allison R. Greenplate
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Immune Health™, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Cécile Alanio
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Mohamed Abdel-Hakeem
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Derek A. Oldridge
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Josephine R. Giles
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Jennifer E. Wu
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Zeyu Chen
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Yinghui Jane Huang
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Jonathan Belman
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Ajinkya Pattekar
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Immune Health™, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Sasikanth Manne
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Oliva Kuthuru
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Jeanette Dougherty
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Brittany Weiderhold
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - Ariel R. Weisman
- Division of Pulmonary, Allergy and Critical Care Medicine, Center for Translational Lung Biology, Lung Biology Institute, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Caroline A. G. Ittner
- Division of Pulmonary, Allergy and Critical Care Medicine, Center for Translational Lung Biology, Lung Biology Institute, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Sigrid Gouma
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Debora Dunbar
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Ian Frank
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Alexander C. Huang
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Laura A. Vella
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - John P. Reilly
- Division of Pulmonary, Allergy and Critical Care Medicine, Center for Translational Lung Biology, Lung Biology Institute, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Scott E. Hensley
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Lubica Rauova
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Liang Zhao
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - Nuala J. Meyer
- Division of Pulmonary, Allergy and Critical Care Medicine, Center for Translational Lung Biology, Lung Biology Institute, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Mortimer Poncz
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Charles S. Abrams
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - E. John Wherry
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Immune Health™, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
9
|
Monteleone JPR, Gao X, Kleijn HJ, Bellanti F, Pelto R. Eculizumab Pharmacokinetics and Pharmacodynamics in Patients With Generalized Myasthenia Gravis. Front Neurol 2021; 12:696385. [PMID: 34795626 PMCID: PMC8594444 DOI: 10.3389/fneur.2021.696385] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 08/20/2021] [Indexed: 11/17/2022] Open
Abstract
Objective: To investigate the pharmacokinetics, pharmacodynamics, and exposure-response of the approved 900/1,200 mg dosing regimen for the terminal complement component 5 (C5) inhibitor eculizumab in patients with generalized myasthenia gravis (gMG). Methods: The analysis used data from 62 patients aged ≥ 18 years with anti-acetylcholine receptor (AChR) antibody-positive refractory gMG who received eculizumab during the REGAIN study (ClinicalTrials.gov: NCT01997229). One- and two-compartment population-pharmacokinetic models were evaluated, and the impact of covariates on pharmacokinetic parameters was assessed. Relationships between eculizumab exposure and free C5 concentration, in vitro hemolytic activity, clinical response, and tolerability were characterized. Results: Steady-state serum eculizumab concentrations were achieved by Week 4 and were sustained throughout the 26-week treatment period. The eculizumab pharmacokinetic data were well-described by a two-compartment model with first-order elimination, including effects of body weight on pharmacokinetic parameters and plasma-exchange events on clearance. Complete inhibition of terminal complement was achieved in nearly all patients at the time of trough and peak eculizumab concentrations at all post-dose timepoints assessed (free C5 < 0.5 μg/ml in 92% of patients; in vitro hemolysis < 20% in 87% of patients). Serum eculizumab concentrations of ≥116 μg/ml achieved free C5 concentrations of < 0.5 μg/ml. Clinical efficacy and tolerability were consistent across the eculizumab exposure range. Conclusions: Rigorous, quantitative, model-based exposure-response analysis of serum eculizumab concentration and response data demonstrated that the approved eculizumab dosing (900/1,200 mg) for adults with anti-AChR antibody-positive refractory gMG rapidly achieved complete inhibition of terminal complement activation and provided sustained clinical efficacy across the eculizumab exposure range.
Collapse
Affiliation(s)
- Jonathan P. R. Monteleone
- Department of Pharmacometrics, PK/PD M&S, Clinical Development and Translational Sciences, Alexion Pharmaceuticals Inc., Boston, MA, United States
| | - Xiang Gao
- Department of Pharmacometrics, PK/PD M&S, Clinical Development and Translational Sciences, Alexion Pharmaceuticals Inc., Boston, MA, United States
| | | | | | - Ryan Pelto
- Department of Pharmacometrics, PK/PD M&S, Clinical Development and Translational Sciences, Alexion Pharmaceuticals Inc., Boston, MA, United States
| |
Collapse
|
10
|
Tammam SN, El Safy S, Ramadan S, Arjune S, Krakor E, Mathur S. Repurpose but also (nano)-reformulate! The potential role of nanomedicine in the battle against SARS-CoV2. J Control Release 2021; 337:258-284. [PMID: 34293319 PMCID: PMC8289726 DOI: 10.1016/j.jconrel.2021.07.028] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 07/15/2021] [Accepted: 07/15/2021] [Indexed: 02/06/2023]
Abstract
The coronavirus disease-19 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) has taken the world by surprise. To date, a worldwide approved treatment remains lacking and hence in the context of rapid viral spread and the growing need for rapid action, drug repurposing has emerged as one of the frontline strategies in the battle against SARS-CoV2. Repurposed drugs currently being evaluated against COVID-19 either tackle the replication and spread of SARS-CoV2 or they aim at controlling hyper-inflammation and the rampaged immune response in severe disease. In both cases, the target for such drugs resides in the lungs, at least during the period where treatment could still provide substantial clinical benefit to the patient. Yet, most of these drugs are administered systemically, questioning the percentage of administered drug that actually reaches the lung and as a consequence, the distribution of the remainder of the dose to off target sites. Inhalation therapy should allow higher concentrations of the drug in the lungs and lower concentrations systemically, hence providing a stronger, more localized action, with reduced adverse effects. Therefore, the nano-reformulation of the repurposed drugs for inhalation is a promising approach for targeted drug delivery to lungs. In this review, we critically analyze, what nanomedicine could and ought to do in the battle against SARS-CoV2. We start by a brief description of SARS-CoV2 structure and pathogenicity and move on to discuss the current limitations of repurposed antiviral and immune-modulating drugs that are being clinically investigated against COVID-19. This account focuses on how nanomedicine could address limitations of current therapeutics, enhancing the efficacy, specificity and safety of such drugs. With the appearance of new variants of SARS-CoV2 and the potential implication on the efficacy of vaccines and diagnostics, the presence of an effective therapeutic solution is inevitable and could be potentially achieved via nano-reformulation. The presence of an inhaled nano-platform capable of delivering antiviral or immunomodulatory drugs should be available as part of the repertoire in the fight against current and future outbreaks.
Collapse
Affiliation(s)
- Salma N Tammam
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, The German University in Cairo (GUC), 11835 Cairo, Egypt.
| | - Sara El Safy
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, The German University in Cairo (GUC), 11835 Cairo, Egypt
| | - Shahenda Ramadan
- Department of Pharmaceutical Technology, Faculty of Pharmacy & Biotechnology, The German University in Cairo (GUC), 11835 Cairo, Egypt
| | - Sita Arjune
- Institute of Biochemistry, Department of Chemistry, Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Eva Krakor
- Institute of Inorganic Chemistry, Department of Chemistry, , University of Cologne, Greinstraße 6, 50939 Cologne, Germany
| | - Sanjay Mathur
- Institute of Inorganic Chemistry, Department of Chemistry, , University of Cologne, Greinstraße 6, 50939 Cologne, Germany
| |
Collapse
|
11
|
Apostolidis SA, Sarkar A, Giannini HM, Goel RR, Mathew D, Suzuki A, Baxter AE, Greenplate AR, Alanio C, Abdel-Hakeem M, Oldridge DA, Giles J, Wu JE, Chen Z, Huang YJ, Pattekar A, Manne S, Kuthuru O, Dougherty J, Weiderhold B, Weisman AR, Ittner CAG, Gouma S, Dunbar D, Frank I, Huang AC, Vella LA, The UPenn COVID Processing Unit, Reilly JP, Hensley SE, Rauova L, Zhao L, Meyer NJ, Poncz M, Abrams CS, Wherry EJ. Signaling through FcγRIIA and the C5a-C5aR pathway mediates platelet hyperactivation in COVID-19. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2021:2021.05.01.442279. [PMID: 33972943 PMCID: PMC8109205 DOI: 10.1101/2021.05.01.442279] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Patients with COVID-19 present with a wide variety of clinical manifestations. Thromboembolic events constitute a significant cause of morbidity and mortality in patients infected with SARS-CoV-2. Severe COVID-19 has been associated with hyperinflammation and pre-existing cardiovascular disease. Platelets are important mediators and sensors of inflammation and are directly affected by cardiovascular stressors. In this report, we found that platelets from severely ill, hospitalized COVID-19 patients exhibit higher basal levels of activation measured by P-selectin surface expression, and have a poor functional reserve upon in vitro stimulation. Correlating clinical features to the ability of plasma from COVID-19 patients to stimulate control platelets identified ferritin as a pivotal clinical marker associated with platelet hyperactivation. The COVID-19 plasma-mediated effect on control platelets was highest for patients that subsequently developed inpatient thrombotic events. Proteomic analysis of plasma from COVID-19 patients identified key mediators of inflammation and cardiovascular disease that positively correlated with in vitro platelet activation. Mechanistically, blocking the signaling of the FcγRIIa-Syk and C5a-C5aR pathways on platelets, using antibody-mediated neutralization, IgG depletion or the Syk inhibitor fostamatinib, reversed this hyperactivity driven by COVID-19 plasma and prevented platelet aggregation in endothelial microfluidic chamber conditions, thus identifying these potentially actionable pathways as central for platelet activation and/or vascular complications in COVID-19 patients. In conclusion, we reveal a key role of platelet-mediated immunothrombosis in COVID-19 and identify distinct, clinically relevant, targetable signaling pathways that mediate this effect. These studies have implications for the role of platelet hyperactivation in complications associated with SARS-CoV-2 infection. COVER ILLUSTRATION ONE-SENTENCE SUMMARY The FcγRIIA and C5a-C5aR pathways mediate platelet hyperactivation in COVID-19.
Collapse
Affiliation(s)
- Sokratis A. Apostolidis
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Rheumatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Amrita Sarkar
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Heather M. Giannini
- Division of Pulmonary, Allergy and Critical Care Medicine, Center for Translational Lung Biology, Lung Biology Institute, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Rishi R. Goel
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Divij Mathew
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Aae Suzuki
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Amy E. Baxter
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Allison R. Greenplate
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Cécile Alanio
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mohamed Abdel-Hakeem
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Derek A. Oldridge
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Josephine Giles
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jennifer E. Wu
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Zeyu Chen
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Yinghui Jane Huang
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ajinkya Pattekar
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sasikanth Manne
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Oliva Kuthuru
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jeanette Dougherty
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Brittany Weiderhold
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Ariel R. Weisman
- Division of Pulmonary, Allergy and Critical Care Medicine, Center for Translational Lung Biology, Lung Biology Institute, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Caroline A. G. Ittner
- Division of Pulmonary, Allergy and Critical Care Medicine, Center for Translational Lung Biology, Lung Biology Institute, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sigrid Gouma
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Debora Dunbar
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ian Frank
- Division of Infectious Diseases, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander C. Huang
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Laura A. Vella
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Division of Infectious Diseases, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - John P. Reilly
- Division of Pulmonary, Allergy and Critical Care Medicine, Center for Translational Lung Biology, Lung Biology Institute, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Scott E. Hensley
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Microbiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Lubica Rauova
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Liang Zhao
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Nuala J. Meyer
- Division of Pulmonary, Allergy and Critical Care Medicine, Center for Translational Lung Biology, Lung Biology Institute, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mortimer Poncz
- Division of Hematology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Charles S. Abrams
- Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - E. John Wherry
- Institute for Immunology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Immune Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Parker Institute for Cancer Immunotherapy, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
12
|
Abstract
The terminal complement protein (C5) inhibitor eculizumab (Soliris®) is the first agent to be specifically approved in the EU, USA, Canada and Japan for the treatment of neuromyelitis optica spectrum disorder (NMOSD) in adults who are aquaporin-4 water channel autoantibody (AQP4-IgG) seropositive and (in the EU only) for those with a relapsing course of disease. In the phase III PREVENT trial, eculizumab significantly reduced the risk of adjudicated relapse relative to placebo in patients with AQP4-IgG-seropositive NMOSD, approximately a quarter of whom did not receive concomitant immunosuppressive therapies. The beneficial effect of eculizumab was seen across all patient subgroups analysed and was accompanied by improvements in neurological and functional disability assessments, as well as generic health-related quality of life measures; it was sustained through 4 years of treatment, according to combined data from the PREVENT trial and an interim analysis of its ongoing open-label extension study. The safety profile of eculizumab in AQP4-IgG-seropositive NMOSD was consistent with that seen for the drug in other approved indications. Thus, eculizumab provides an effective, generally well tolerated and approved treatment option for this rare, disabling and potentially life-threatening condition.
Collapse
|
13
|
Characteristics of and meningococcal disease prevention strategies for commercially insured persons receiving eculizumab in the United States. PLoS One 2020; 15:e0241989. [PMID: 33180804 PMCID: PMC7660549 DOI: 10.1371/journal.pone.0241989] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 10/25/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Eculizumab is a licensed treatment for several rare, complement-mediated diseases. Eculizumab use is associated with an approximately 2,000-fold increased meningococcal disease risk. In the United States, meningococcal vaccines are recommended for eculizumab recipients but there are no recommendations on use of long-term antibiotic prophylaxis. We describe characteristics of and meningococcal vaccine and antibiotic receipt in U.S. eculizumab recipients to inform meningococcal disease prevention strategies. METHODS Persons in the IBM® MarketScan® Research Databases with ≥1 claim for eculizumab injection during 2007-2017 were included. Indication for eculizumab use, meningococcal vaccine receipt, and antibiotic receipt were assessed using International Classification of Diseases-9/10 diagnosis codes, vaccine administration procedure codes, and antibiotic codes from pharmacy claims, respectively. RESULTS Overall 696 persons met the inclusion criteria. Paroxysmal nocturnal hemoglobinuria (PNH) and atypical hemolytic uremic syndrome (aHUS) were the most common indications for eculizumab use (41% and 37%, respectively); 20% had an undetermined indication. From June 2015 through December 2017, 28% (41/148) of continuously-enrolled patients received ≥1 serogroup B vaccine dose. For serogroup ACWY conjugate vaccine, 45% (91/201) of patients received ≥1 dose within five years of their most recent eculizumab dose, as recommended. Of eculizumab recipients with outpatient prescription data, 7% (41/579) received antibiotics for ≥50% of the period of increased risk for meningococcal disease. CONCLUSION Many eculizumab recipients had an undetermined indication for eculizumab use; few were up-to-date for recommended meningococcal vaccines or were prescribed antibiotics long-term. These findings can inform further investigation of how to best protect this population from meningococcal disease.
Collapse
|
14
|
Bordron A, Bagacean C, Tempescul A, Berthou C, Bettacchioli E, Hillion S, Renaudineau Y. Complement System: a Neglected Pathway in Immunotherapy. Clin Rev Allergy Immunol 2020; 58:155-171. [PMID: 31144209 DOI: 10.1007/s12016-019-08741-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Approved for the treatment of autoimmune diseases, hematological malignancies, and solid cancers, several monoclonal antibodies (mAb) make use of complement in their mechanism of action. Such an assessment is based on comprehensive investigations that used mouse models, in vitro studies, and analyses from patients at initiation (basal level to highlight deficiencies) and after treatment initiation (mAb impact on complement), which have further provided key insights into the importance of the complement activation and/or complement deficiencies in mAb activity. Accordingly, new approaches can now be developed with the final objective of increasing the clinical efficacy of mAb. These improvements include (i) the concurrent administration of fresh frozen plasma during mAb therapy; (ii) mAb modifications such as immunoglobulin G subclass switching, Fc mutation, or IgG hexamerization to improve the fixation and activation of C1q; (iii) optimization of the target recognition to induce a higher complement-dependent cytotoxicity (CDC) and/or complement-dependant cellular cytotoxicity (CDCC); and (iv) the control of soluble and cellular complement inhibitors.
Collapse
Affiliation(s)
- Anne Bordron
- Inserm UMR1227, B lymphocytes and autoimmunity, University of Brest, Brest, France
| | - Cristina Bagacean
- Inserm UMR1227, B lymphocytes and autoimmunity, University of Brest, Brest, France.,Service d'Hématologie, CHU de Brest, Brest, France
| | - Adrian Tempescul
- Inserm UMR1227, B lymphocytes and autoimmunity, University of Brest, Brest, France.,Service d'Hématologie, CHU de Brest, Brest, France
| | - Christian Berthou
- Inserm UMR1227, B lymphocytes and autoimmunity, University of Brest, Brest, France.,Service d'Hématologie, CHU de Brest, Brest, France
| | | | - Sophie Hillion
- Inserm UMR1227, B lymphocytes and autoimmunity, University of Brest, Brest, France.,Laboratory of Immunology and Immunotherapy, CHU de Brest, Brest, France
| | - Yves Renaudineau
- Inserm UMR1227, B lymphocytes and autoimmunity, University of Brest, Brest, France. .,Laboratory of Immunology and Immunotherapy, CHU de Brest, Brest, France.
| |
Collapse
|
15
|
Draskovic B, Nikolic T, Jacovic S, Petrovic D. Acute Kidney Damage in Pregnancy: Etiopathogenesis, Diagnostics and Basic Principles of Treatment. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2020. [DOI: 10.1515/sjecr-2017-0058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Acute kidney damage associated with pregnancy occurs in 1/20.000 pregnancies. In developing countries, the main cause of the development of acute kidney damage is septic abortion, and preeclampsia in the developed countries of the world. Preeclampsia is defined as newly developed hypertension, proteinuria and swelling in pregnant women after the 20th week of gestation. It occurs due to disorders in the development of placenta and systemic disorders of the function of the endothelium of the mother. It is treated with methyldopa, magnesium sulfate and timely delivery. Urgent delivery is indicated if the age of gestation is ≥ 34 weeks. HELLP syndrome is a difficult form of preeclampsia. Its main characteristics are decreased platelet count, microangiopathic hemolysis anemia, increased concentration of aminotransferase in the serum and acute kidney damage. Severe HELLP syndrome is treated with emergency delivery, antihypertensives, magnesium sulfate, and in some cases plasmapheresis and hemodialysis. Acute fatty liver in pregnancy occurs because of decreased activity of the LCHAD enzyme of the fetus. Due to the reduced beta oxidation of fatty acids in the hepatocytes of the fetus, long chain fatty acids that cause damage to the mother’s hepatocytes are released. Swansea criteria are used for diagnosis, and the difficult form of the disease is treated with plasmapheresis and extracorporeal liver support. Atypical HUS is due to a reduced protein activity that regulates the activity of the alternative pathway of the complement system. Its main features are thrombocytopenia, microangiopathic hemolytic anemia and acute kidney damage. It is treated with plasmapheresis, and in case of resistance with eculizumab. Thrombotic thrombocytopenic purpura is due to decreased activity of the ADAMTS13 enzyme. It is characterized by thrombocytopenia, microangiopathic hemolytic anemia, high temperature, nervous system disorders and acute kidney damage. It is treated with plasmapheresis, and severe form of disease with corticosteroids and azathioprine. Early detection and timely treatment of acute kidney damage provides a good outcome for the mother and fetus.
Collapse
Affiliation(s)
- Branislava Draskovic
- Clinic of Urology, Nephrology and Dialysis, Clinical center Kragujevac , Kragujevac , Serbia
| | - Tomislav Nikolic
- Clinic of Urology, Nephrology and Dialysis, Clinical center Kragujevac , Kragujevac , Serbia
- University of Kragujevac , Faculty of Medical Sciences , Kragujevac , Serbia
| | - Sasa Jacovic
- Medicines and Medical Devices Agency of Serbia , Belgrade , Serbia
| | - Dejan Petrovic
- Clinic of Urology, Nephrology and Dialysis, Clinical center Kragujevac , Kragujevac , Serbia
- University of Kragujevac , Faculty of Medical Sciences , Kragujevac , Serbia
| |
Collapse
|
16
|
Abstract
The humanized monoclonal antibody eculizumab (Soliris®) is a complement inhibitor indicated for use in anti-acetylcholine receptor (AChR) antibody-positive adults with generalized myasthenia gravis (gMG) in the USA, refractory gMG in the EU, or gMG with symptoms that are difficult to control with high-dose IVIg therapy or PLEX in Japan. It is the first complement inhibitor to be approved for use in these patients. In the well-designed, 26-week REGAIN study in patients with anti-AChR-positive refractory gMG, although a statistically significant benefit of eculizumab over placebo in the prespecified primary endpoint analysis (change from baseline in MG-activities of daily living (ADL) score assessed by worst-rank ANCOVA) was not formally demonstrated, preplanned and post hoc sensitivity analyses of this outcome, as well as other secondary outcomes supported the efficacy of eculizumab. Overall, patients receiving eculizumab experienced significant improvements in the ADL, muscle strength and health-related quality of life (HR-QOL) parameters relative to patients receiving placebo. Moreover, an ongoing extension of REGAIN showed that treatment benefits with eculizumab were sustained during continued therapy for at least 52 weeks. Eculizumab was generally well tolerated in these studies, with a tolerability profile similar to that reported previously in other indications. Although several questions remain, such as duration of treatment, cost effectiveness and long-term efficacy and tolerability, current evidence indicates that eculizumab is a valuable emerging therapy for patients with refractory gMG.
Collapse
Affiliation(s)
- Sohita Dhillon
- Springer, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| |
Collapse
|
17
|
|
18
|
Neisseria cinerea bacteremia in a patient receiving eculizumab: a case report. Infection 2017; 46:271-274. [DOI: 10.1007/s15010-017-1090-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 10/23/2017] [Indexed: 10/18/2022]
|
19
|
Parikh SR, Lucidarme J, Bingham C, Warwicker P, Goodship T, Borrow R, Ladhani SN. Meningococcal B Vaccine Failure With a Penicillin-Resistant Strain in a Young Adult on Long-Term Eculizumab. Pediatrics 2017; 140:peds.2016-2452. [PMID: 28864711 DOI: 10.1542/peds.2016-2452] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/03/2017] [Indexed: 11/24/2022] Open
Abstract
We describe a case of invasive meningococcal disease due to a vaccine-preventable and penicillin-resistant strain in a fully immunized young adult on long-term complement inhibitor therapy and daily penicillin chemoprophylaxis. Eculizumab is a humanized monoclonal antibody that binds human complement C5 protein and inhibits the terminal complement pathway. It is currently recommended for the treatment of complement-mediated thrombotic microangiopathies. An unwanted complication of inhibiting complement, however, is an increased risk of invasive meningococcal disease. Here, we report the first case of meningococcal group B vaccine failure in a young adult receiving eculizumab for atypical hemolytic uremic syndrome. She developed invasive meningococcal disease due to a vaccine-preventable and penicillin-resistant meningococcal group B strain 4 months after receiving 2 doses of meningococcal group B vaccine while on oral penicillin prophylaxis against meningococcal infection.
Collapse
Affiliation(s)
- Sydel R Parikh
- Immunisation, Hepatitis, and Blood Safety Department, Public Health England, London, United Kingdom
| | - Jay Lucidarme
- Vaccine Evaluation Unit, Public Health England, Manchester, United Kingdom
| | - Coralie Bingham
- Exeter Kidney Unit, Royal Devon and Exeter Hospital, Devon, United Kingdom
| | - Paul Warwicker
- Lister Renal Unit, Lister Hospital, Hertfordshire, United Kingdom
| | - Tim Goodship
- Institute of Human Genetics, Newcastle University, Newcastle, United Kingdom; and
| | - Ray Borrow
- Vaccine Evaluation Unit, Public Health England, Manchester, United Kingdom
| | - Shamez N Ladhani
- Immunisation, Hepatitis, and Blood Safety Department, Public Health England, London, United Kingdom; .,St George's University of London, London, United Kingdom
| |
Collapse
|
20
|
Konar M, Granoff DM. Eculizumab treatment and impaired opsonophagocytic killing of meningococci by whole blood from immunized adults. Blood 2017; 130. [PMID: 28630122 PMCID: PMC5561903 DOI: 10.1182/blood-2017-05-781450] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Eculizumab, a humanized anti-complement C5 monoclonal antibody (mAb) for treatment of paroxysmal nocturnal hemoglobinuria (PNH) and atypical hemolytic uremic syndrome, blocks the terminal complement pathway required for serum bactericidal activity (SBA). Because treated patients are at >1000-fold increased risk of meningococcal disease, vaccination is recommended; whether vaccination can protect by opsonophagocytic activity in the absence of SBA is not known. Meningococci were added to anticoagulated blood from 12 healthy adults vaccinated with meningococcal serogroup B and serogroup A, C, W, Y vaccines. Bacterial survival was measured after 3-hour incubation in the presence of eculizumab or control complement factor D inhibitor ACH-4471, which blocks the complement alternative pathway (AP) and is in phase 2 development for treatment of PNH. In the absence of inhibitors, colony formation units (CFUs) per milliliter in blood from all 12 immunized subjects decreased from ∼4000 at time 0 to sterile cultures at 3 hours. In the presence of eculizumab, there was a >22-fold increase in geometric mean CFUs per milliliter (90 596 and 114 683 CFU/mL for serogroup B and C strains, respectively; P < .0001 compared with time 0). In the presence of ACH-4471, there was a >12-fold decrease (23 and 331 CFU/mL, respectively; P < .0001). The lack of meningococci killing by blood containing eculizumab resulted from inhibition of release of C5a, a C5 split product needed for upregulation of phagocytosis. The results provide an explanation for the large number of cases of meningococcal disease in immunized patients being treated with eculizumab and suggest that vaccination may provide better protection against meningococcal disease in patients treated with an AP-specific inhibitor.
Collapse
Affiliation(s)
- Monica Konar
- Center for Immunobiology and Vaccine Development, UCSF Benioff Children's Hospital Oakland, Oakland, CA
| | - Dan M Granoff
- Center for Immunobiology and Vaccine Development, UCSF Benioff Children's Hospital Oakland, Oakland, CA
| |
Collapse
|
21
|
Macia M, de Alvaro Moreno F, Dutt T, Fehrman I, Hadaya K, Gasteyger C, Heyne N. Current evidence on the discontinuation of eculizumab in patients with atypical haemolytic uraemic syndrome. Clin Kidney J 2016. [PMID: 28621343 PMCID: PMC5466111 DOI: 10.1093/ckj/sfw115] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Background. Atypical haemolytic uraemic syndrome (aHUS) is a rare, life-threatening disorder for which eculizumab is the only approved treatment. Life-long treatment is indicated; however, eculizumab discontinuation has been reported. Methods. Unpublished authors' cases and published cases of eculizumab discontinuation are reviewed. We also report eculizumab discontinuation data from five clinical trials, plus long-term extensions and the global aHUS Registry. Results. Of six unpublished authors' cases, four patients had a subsequent thrombotic microangiopathy (TMA) manifestation within 12 months of discontinuation. Case reports of 52 patients discontinuing eculizumab were identified; 16 (31%) had a subsequent TMA manifestation. In eculizumab clinical trials, 61/130 patients discontinued treatment between 2008 and 2015. Median follow-up post-discontinuation was 24 weeks and during this time 12 patients experienced 15 severe TMA complications and 9 of the 12 patients restarted eculizumab. TMA complications occurred irrespective of identified genetic mutation, high risk polymorphism or auto-antibody. In the global aHUS Registry, 76/296 patients (26%) discontinued, 12 (16%) of whom restarted. Conclusions. The currently available evidence suggests TMA manifestations following discontinuation are unpredictable in both severity and timing. For evidence-based decision making, better risk stratification and valid monitoring strategies are required. Until these exist, the risk versus benefit of eculizumab discontinuation, either in specific clinical situations or at selected time points, should include consideration of the risk of further TMA manifestations.
Collapse
Affiliation(s)
- Manuel Macia
- Nephrology service, University Hospital NS de Candelaria, Santa Cruz de Tenerife, Spain
| | | | - Tina Dutt
- Roald Dahl Haemostasis and Thrombosis Centre, Royal Liverpool University Hospital, Liverpool, UK
| | - Ingela Fehrman
- Division of Transplant Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Karine Hadaya
- Divisions of Nephrology and Transplantation, Geneva University Hospitals, Geneva, Switzerland
| | | | - Nils Heyne
- Department of Endocrinology and Diabetology, Angiology, Nephrology and Clinical Chemistry, University of Tübingen, Tübingen, Germany
| |
Collapse
|
22
|
van Greevenbroek MMJ, Schalkwijk CG, Stehouwer CDA. Dysfunctional adipose tissue and low-grade inflammation in the management of the metabolic syndrome: current practices and future advances. F1000Res 2016; 5. [PMID: 27803798 PMCID: PMC5070595 DOI: 10.12688/f1000research.8971.1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/10/2016] [Indexed: 12/12/2022] Open
Abstract
The ongoing worldwide obesity epidemic makes the metabolic syndrome an increasingly important entity. In this review, we provide a short background on the metabolic syndrome, we discuss recent developments in the three main options that have been identified for intervention in the metabolic syndrome, i.e. lifestyle and surgical and pharmacological interventions, and we focus on different views in the literature and also include our own viewpoints on the metabolic syndrome. In addition, we discuss some emerging treatment targets for adipose tissue dysfunction and low-grade inflammation, i.e. activation of the inflammasome and the complement system, and consider some selected opportunities for intervention in these processes.
Collapse
Affiliation(s)
- Marleen M J van Greevenbroek
- Maastricht University Medical Center, Maastricht, 6229 ER, Netherlands; CARIM School for Cardiovascular Diseases, Maastricht, 6229 ER, Netherlands
| | - Casper G Schalkwijk
- Maastricht University Medical Center, Maastricht, 6229 ER, Netherlands; CARIM School for Cardiovascular Diseases, Maastricht, 6229 ER, Netherlands
| | - Coen D A Stehouwer
- Maastricht University Medical Center, Maastricht, 6229 ER, Netherlands; CARIM School for Cardiovascular Diseases, Maastricht, 6229 ER, Netherlands; Academic Hospital Maastricht, Maastricht, 6229 HX, Netherlands
| |
Collapse
|
23
|
Hess C, Kemper C. Complement-Mediated Regulation of Metabolism and Basic Cellular Processes. Immunity 2016; 45:240-54. [PMID: 27533012 PMCID: PMC5019180 DOI: 10.1016/j.immuni.2016.08.003] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/25/2016] [Accepted: 08/01/2016] [Indexed: 02/07/2023]
Abstract
Complement is well appreciated as a critical arm of innate immunity. It is required for the removal of invading pathogens and works by directly destroying them through the activation of innate and adaptive immune cells. However, complement activation and function is not confined to the extracellular space but also occurs within cells. Recent work indicates that complement activation regulates key metabolic pathways and thus can impact fundamental cellular processes, such as survival, proliferation, and autophagy. Newly identified functions of complement include a key role in shaping metabolic reprogramming, which underlies T cell effector differentiation, and a role as a nexus for interactions with other effector systems, in particular the inflammasome and Notch transcription-factor networks. This review focuses on the contributions of complement to basic processes of the cell, in particular the integration of complement with cellular metabolism and the potential implications in infection and other disease settings.
Collapse
Affiliation(s)
- Christoph Hess
- Department of Biomedicine, Immunobiology, University of Basel, 20 Hebelstrasse, 4031 Basel, Switzerland.
| | - Claudia Kemper
- Division of Transplant Immunology and Mucosal Biology, Medical Reseaerch Council Centre for Transplantation, King's College London, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK; Laboratory of Molecular Immunology and the Immunology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
24
|
Jin A, Boroujerdi-Rad L, Shah G, Chen JLT. Thrombotic microangiopathy and human immunodeficiency virus in the era of eculizumab. Clin Kidney J 2016; 9:576-9. [PMID: 27478600 PMCID: PMC4957717 DOI: 10.1093/ckj/sfw035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 03/31/2016] [Indexed: 01/16/2023] Open
Abstract
Thrombotic microangiopathies (TMAs) include thrombotic thromobocytopenic purpura and hemolytic uremic syndrome (HUS). Among these conditions, atypical HUS is now recognized to be a disease of alternative complement pathway dysregulation. Eculizumab is a recombinant humanized monoclonal antibody that binds to the complement protein C5 and prevents the cleavage of C5 to C5a and C5b. Eculizumab has been used as a novel treatment for complement-mediated TMA. We present a case of a patient with human immunodeficiency virus infection who developed TMA and was successfully treated with eculizumab. The effect of long-term treatment with this new medication is unknown, and further studies are needed to establish guidelines in the management of this condition.
Collapse
Affiliation(s)
- Anna Jin
- Division of Nephrology and Hypertension, Department of Medicine, University of California Irvine, Orange, CA, USA; Nephrology Section, Department of Medicine, Long Beach Veteran Affairs Health System, Long Beach, CA, USA
| | - Laleh Boroujerdi-Rad
- Division of Nephrology and Hypertension, Department of Medicine, University of California Irvine, Orange, CA, USA; Nephrology Section, Department of Medicine, Long Beach Veteran Affairs Health System, Long Beach, CA, USA
| | - Gaurang Shah
- Division of Nephrology and Hypertension, Department of Medicine, University of California Irvine, Orange, CA, USA; Nephrology Section, Department of Medicine, Long Beach Veteran Affairs Health System, Long Beach, CA, USA
| | - Joline L T Chen
- Division of Nephrology and Hypertension, Department of Medicine, University of California Irvine, Orange, CA, USA; Nephrology Section, Department of Medicine, Long Beach Veteran Affairs Health System, Long Beach, CA, USA
| |
Collapse
|
25
|
Improved renal recovery in patients with atypical hemolytic uremic syndrome following rapid initiation of eculizumab treatment. J Nephrol 2016; 30:127-134. [PMID: 26995002 PMCID: PMC5316393 DOI: 10.1007/s40620-016-0288-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 02/13/2016] [Indexed: 11/30/2022]
Abstract
Background Eculizumab is approved for atypical hemolytic uremic syndrome (aHUS). Guidelines discuss the importance of prompt treatment. We report a post hoc analysis investigating the effect of baseline factors, including patient characteristics and time from the latest aHUS manifestation to eculizumab initiation, on change from baseline in estimated glomerular filtration rate (eGFR) and other outcomes. Methods Data were pooled from four phase 2, open-label, single-arm, prospective clinical studies of eculizumab for patients with aHUS. Multivariate regressions identified predictors of eGFR change from baseline. The proportion of patients achieving sustained eGFR increase (defined: ≥15 ml/min/1.73 m2 for ≥28 days) and platelet count normalization were evaluated 1 year post-treatment. Baseline characteristics and eGFR outcomes were summarized by time to treatment from last aHUS manifestation [≤7 days (n = 21) versus >7 days (n = 76)]. Results Baseline eGFR were similar between groups. Multivariate regression analysis demonstrated time from aHUS manifestation to eculizumab treatment, age, baseline lactate dehydrogenase (LDH) and baseline hemoglobin were independently predictive of eGFR change from baseline. Mean eGFR change from baseline at 1 year was significantly higher in patients treated in ≤7 days than >7 days (57 vs. 23 ml/min/1.73 m2, p = 0.0098). After 1 year, 17/21 and 36/76 patients in the ≤7 and >7 day groups, respectively, achieved a sustained increase in eGFR. Mean time to platelet count normalization was similar between groups. Conclusions Younger age, higher baseline LDH and lower baseline hemoglobin were associated with greater eGFR improvements. Early eculizumab initiation led to improved renal recovery, demonstrating the importance of rapid diagnosis and treatment of patients with aHUS. Electronic supplementary material The online version of this article (doi:10.1007/s40620-016-0288-3) contains supplementary material, which is available to authorized users.
Collapse
|
26
|
Orandi AB, Theisen JWM, Saland J, Davis TK. Fever, Jaundice, Abdominal Pain, Skin Lesions, and Dark Urine for 2 Days. Clin Pediatr (Phila) 2016; 55:308-11. [PMID: 26054779 DOI: 10.1177/0009922815589918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
| | | | | | - T Keefe Davis
- Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
27
|
Agudo I, Souto-Moura T, Azevedo L, Cavaco R, Germano N, Bento L. Atypical hemolytic uremic syndrome in intensive care: Case report in an adult. Med Intensiva 2015; 40:188-90. [PMID: 26596222 DOI: 10.1016/j.medin.2015.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2015] [Revised: 08/29/2015] [Accepted: 09/15/2015] [Indexed: 11/27/2022]
Affiliation(s)
- I Agudo
- Unidade de Urgência Médica, Hospital de São José, Centro Hospitalar de Lisboa Central, EPE, Lisboa, Portugal
| | - T Souto-Moura
- Unidade de Urgência Médica, Hospital de São José, Centro Hospitalar de Lisboa Central, EPE, Lisboa, Portugal.
| | - L Azevedo
- Unidade de Urgência Médica, Hospital de São José, Centro Hospitalar de Lisboa Central, EPE, Lisboa, Portugal
| | - R Cavaco
- Unidade de Urgência Médica, Hospital de São José, Centro Hospitalar de Lisboa Central, EPE, Lisboa, Portugal
| | - N Germano
- Unidade de Urgência Médica, Hospital de São José, Centro Hospitalar de Lisboa Central, EPE, Lisboa, Portugal
| | - L Bento
- Unidade de Urgência Médica, Hospital de São José, Centro Hospitalar de Lisboa Central, EPE, Lisboa, Portugal
| |
Collapse
|
28
|
Ferreira E, Oliveira N, Marques M, Francisco L, Santos A, Carreira A, Campos M. Eculizumab for the treatment of an atypical hemolytic uremic syndrome with mutations in complement factor I and C3. Nefrologia 2015; 36:72-5. [PMID: 26541438 DOI: 10.1016/j.nefro.2015.07.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 05/27/2015] [Accepted: 07/29/2015] [Indexed: 11/16/2022] Open
Affiliation(s)
- Emanuel Ferreira
- Servicio de Nefrología, Centro Hospitalar e Universitário de Coimbra-Hospital Geral, Coimbra, Portugal.
| | - Nuno Oliveira
- Servicio de Nefrología, Centro Hospitalar e Universitário de Coimbra-Hospital Geral, Coimbra, Portugal
| | - Maria Marques
- Servicio de Nefrología, Centro Hospitalar e Universitário de Coimbra-Hospital Geral, Coimbra, Portugal
| | - Luís Francisco
- Servicio de Hematología, Centro Hospitalar e Universitário de Coimbra-Hospital Geral, Coimbra, Portugal
| | - Ana Santos
- Servicio de Nefrología, Centro Hospitalar e Universitário de Coimbra-Hospital Geral, Coimbra, Portugal
| | - Armando Carreira
- Servicio de Nefrología, Centro Hospitalar e Universitário de Coimbra-Hospital Geral, Coimbra, Portugal
| | - Mário Campos
- Servicio de Nefrología, Centro Hospitalar e Universitário de Coimbra-Hospitais da Universidade de Coimbra, Coimbra, Portugal
| |
Collapse
|
29
|
Jodele S, Fukuda T, Mizuno K, Vinks AA, Laskin BL, Goebel J, Dixon BP, Chima RS, Hirsch R, Teusink A, Lazear D, Lane A, Myers KC, Dandoy CE, Davies SM. Variable Eculizumab Clearance Requires Pharmacodynamic Monitoring to Optimize Therapy for Thrombotic Microangiopathy after Hematopoietic Stem Cell Transplantation. Biol Blood Marrow Transplant 2015; 22:307-315. [PMID: 26456258 DOI: 10.1016/j.bbmt.2015.10.002] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Accepted: 10/02/2015] [Indexed: 10/22/2022]
Abstract
Thrombotic microangiopathy (TMA) after hematopoietic stem cell transplantation (HSCT) associated with terminal complement activation, as measured by elevated plasma terminal complement (sC5b-9) concentrations, has a very high mortality. The complement inhibitor eculizumab may be a therapeutic option for HSCT-associated TMA. We examined the pharmacokinetics and pharmacodynamics (PK/PD) of eculizumab in children and young adult HSCT recipients with TMA and activated complement to determine drug dosing requirements for future efficacy trials. We analyzed prospectively collected laboratory samples and clinical data from 18 HSCT recipients with high-risk TMA presenting with complement activation who were treated with eculizumab. We measured eculizumab serum concentrations, total hemolytic complement activity, and plasma sC5b-9 concentrations. Population PK/PD analyses correlated eculizumab concentrations with complement blockade and clinical response and determined interindividual differences in PK parameters. We also compared transplant survival in patients treated with eculizumab (n = 18) with patients with the same high-risk TMA features who did not receive any targeted therapy during a separate prospective observational study (n = 11). In the PK analysis, we found significant interpatient variability in eculizumab clearance, ranging from 16 to 237 mL/hr/70 kg in the induction phase. The degree of complement activation measured by sC5b-9 concentrations at the start of therapy, in addition to actual body weight, was a significant determinant of eculizumab clearance and disease response. Sixty-one percent of treated patients had complete resolution of TMA and were able to safely discontinue eculizumab without disease recurrence. Overall survival was significantly higher in treated subjects compared with untreated patients (56% versus 9%, P = .003). Complement blocking therapy is associated with improved survival in HSCT patients with high-risk TMA who historically have dismal outcomes, but eculizumab pharmacokinetics in HSCT recipients differ significantly from reports in other diseases like atypical hemolytic uremic syndrome and paroxysmal nocturnal hemoglobinuria. Our eculizumab dosing algorithm, including pr-treatment plasma sC5b-9 concentrations, patient's actual body weight, and the first eculizumab dose (mg), accurately determined eculizumab concentration-time profiles for HSCT recipients with high-risk TMA. This algorithm may guide eculizumab treatment and ensure that future efficacy studies use the most clinically appropriate and cost-efficient dosing schedules.
Collapse
Affiliation(s)
- Sonata Jodele
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.
| | - Tsuyoshi Fukuda
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kana Mizuno
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Alexander A Vinks
- Division of Clinical Pharmacology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Benjamin L Laskin
- Division of Nephrology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Jens Goebel
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Bradley P Dixon
- Division of Nephrology and Hypertension, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Ranjit S Chima
- Division of Critical Care, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Russel Hirsch
- Division of Cardiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Ashley Teusink
- Department of Pharmacy, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Danielle Lazear
- Department of Pharmacy, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Adam Lane
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Kasiani C Myers
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Christopher E Dandoy
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Stella M Davies
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
30
|
Bao L, Cunningham PN, Quigg RJ. Complement in Lupus Nephritis: New Perspectives. KIDNEY DISEASES 2015; 1:91-9. [PMID: 27536669 DOI: 10.1159/000431278] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Accepted: 05/06/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is an autoimmune disorder caused by loss of tolerance to self-antigens, the production of autoantibodies and deposition of complement-fixing immune complexes (ICs) in injured tissues. SLE is characterized by a wide range of clinical manifestations and targeted organs, with lupus nephritis being one of the most serious complications. The complement system consists of three pathways and is tightly controlled by a set of regulatory proteins to prevent injudicious complement activation on host tissue. The involvement of the complement system in the pathogenesis of SLE is well accepted; yet, its exact role is still not clear. SUMMARY Complement plays dual roles in the pathogenesis of SLE. On the one hand, the complement system appears to have protective features in that hereditary homozygous deficiencies of classical pathway components, such as C1q and C4, are associated with an increased risk for SLE. On the other hand, IC-mediated activation of complement in affected tissues is clearly evident in both experimental and human SLE along with pathological features that are logical consequences of complement activation. Studies in genetically altered mice have shown that lack of complement inhibitors, such as complement factor H (CFH) or decay-accelerating factor (DAF) accelerates the development of experimental lupus nephritis, while treatment with recombinant protein inhibitors, such as Crry-Ig, CR2-Crry, CR2-DAF and CR2-CFH, ameliorates the disease development. Complement-targeted drugs, including soluble complement receptor 1 (TP10), C1 esterase inhibitor and a monoclonal anti-C5 antibody (eculizumab), have been shown to inhibit complement safely, and are now being investigated in a variety of clinical conditions. KEY MESSAGES SLE is an autoimmune disorder which targets multiple systems. Complement is centrally involved and plays dual roles in the pathogenesis of SLE. Studies from experimental lupus models and clinical trials support the use of complement-targeted therapy in the treatment of SLE.
Collapse
Affiliation(s)
- Lihua Bao
- Section of Nephrology, Department of Medicine, University of Chicago, Chicago, Ill., USA
| | - Patrick N Cunningham
- Section of Nephrology, Department of Medicine, University of Chicago, Chicago, Ill., USA
| | - Richard J Quigg
- Division of Nephrology, University at Buffalo School of Medicine, Buffalo, N.Y., USA
| |
Collapse
|
31
|
Abstract
First identified in human serum in the late 19th century as a 'complement' to antibodies in mediating bacterial lysis, the complement system emerged more than a billion years ago probably as the first humoral immune system. The contemporary complement system consists of nearly 60 proteins in three activation pathways (classical, alternative and lectin) and a terminal cytolytic pathway common to all. Modern molecular biology and genetics have not only led to further elucidation of the structure of complement system components, but have also revealed function-altering rare variants and common polymorphisms, particularly in regulators of the alternative pathway, that predispose to human disease by creating 'hyperinflammatory complement phenotypes'. To treat these 'complementopathies', a monoclonal antibody against the initiator of the membrane attack complex, C5, has received approval for use. Additional therapeutic reagents are on the horizon.
Collapse
Affiliation(s)
- M K Liszewski
- Division of Rheumatology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - J P Atkinson
- Division of Rheumatology, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
32
|
Protective efficacy and pharmacokinetics of human/mouse chimeric anti-Stx1 and anti-Stx2 antibodies in mice. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2015; 22:448-55. [PMID: 25716230 DOI: 10.1128/cvi.00022-15] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In the United States, Shiga toxin (Stx)-producing Escherichia coli (STEC) is the most frequent infectious cause of hemorrhagic colitis. Hemolytic uremic syndrome (HUS) is a serious sequela that may develop after STEC infection that can lead to renal failure and death in up to 10% of cases. STEC can produce one or more types of Stx, Stx1 and/or Stx2, and Stx1 and Stx2 are responsible for HUS-mediated kidney damage. We previously generated two monoclonal antibodies (MAbs) that neutralize the toxicity of Stx1 or Stx2. In this study, we evaluated the protective efficacy of human/mouse chimeric versions of those monoclonal antibodies, named cαStx1 and cαStx2. Mice given an otherwise lethal dose of Stx1 were protected from death when injected with cαStx1 either 1 h before or 1 h after toxin injection. Additionally, streptomycin-treated mice fed the mouse-lethal STEC strain B2F1 that produces the Stx2 variant Stx2d were protected when given a dose of 0.1 mg of cαStx2/kg of body weight administered up to 72 h post-oral bacterial challenge. Since many STEC strains produce both Stx1 and Stx2 and since either toxin may lead to the HUS, we also assessed the protective efficacy of the combined MAbs. We found that both antibodies were required to protect mice from the presence of both Stx1 and Stx2. Pharmacokinetic studies indicated that cαStx1 and cαStx2 had serum half-lives (t1/2) of about 50 and 145 h, respectively. We propose that cαStx1 and cαStx2, both of which have been tested for safety in humans, could be used therapeutically for prevention or treatment early in the development of HUS.
Collapse
|
33
|
Melis JPM, Strumane K, Ruuls SR, Beurskens FJ, Schuurman J, Parren PWHI. Complement in therapy and disease: Regulating the complement system with antibody-based therapeutics. Mol Immunol 2015; 67:117-30. [PMID: 25697848 DOI: 10.1016/j.molimm.2015.01.028] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 01/26/2015] [Accepted: 01/27/2015] [Indexed: 12/23/2022]
Abstract
Complement is recognized as a key player in a wide range of normal as well as disease-related immune, developmental and homeostatic processes. Knowledge of complement components, structures, interactions, and cross-talk with other biological systems continues to grow and this leads to novel treatments for cancer, infectious, autoimmune- or age-related diseases as well as for preventing transplantation rejection. Antibodies are superbly suited to be developed into therapeutics with appropriate complement stimulatory or inhibitory activity. Here we review the design, development and future of antibody-based drugs that enhance or dampen the complement system.
Collapse
Affiliation(s)
| | | | | | | | | | - Paul W H I Parren
- Genmab, Utrecht, The Netherlands; Department of Immunohematology and Blood Transfusion, Leiden University Medical Center, Leiden, The Netherlands.
| |
Collapse
|
34
|
Efficacy and safety of eculizumab in atypical hemolytic uremic syndrome from 2-year extensions of phase 2 studies. Kidney Int 2015; 87:1061-73. [PMID: 25651368 PMCID: PMC4424817 DOI: 10.1038/ki.2014.423] [Citation(s) in RCA: 301] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2013] [Revised: 09/30/2014] [Accepted: 10/30/2014] [Indexed: 12/11/2022]
Abstract
Atypical hemolytic uremic syndrome (aHUS) is a rare, possibly life-threatening disease characterized by platelet activation, hemolysis and thrombotic microangiopathy (TMA) leading to renal and other end-organ damage. We originally conducted two phase 2 studies (26 weeks and 1 year) evaluating eculizumab, a terminal complement inhibitor, in patients with progressing TMA (trial 1) and those with long duration of aHUS and chronic kidney disease (trial 2). The current analysis assessed outcomes after 2 years (median eculizumab exposure 100 and 114 weeks, respectively). At all scheduled time points, eculizumab inhibited terminal complement activity. In trial 1 with 17 patients, the platelet count was significantly improved from baseline, and hematologic normalization was achieved in 13 patients at week 26, and in 15 patients at both 1 and 2 years. The estimated glomerular filtration rate (eGFR) was significantly improved compared with baseline and year 1. In trial 2 with 20 patients, TMA event-free status was achieved by 16 patients at week 26, 17 patients at year 1, and 19 patients at year 2. Criteria for hematologic normalization were met by 18 patients at each time point. Improvement of 15 ml/min per 1.73 m2 or more in eGFR was achieved by 1 patient at week 26, 3 patients at 1 year, and 8 patients at 2 years. The mean change in eGFR was not significant compared with baseline, week 26, or year 1. Eculizumab was well tolerated, with no new safety concerns or meningococcal infections. Thus, a 2-year analysis found that the earlier clinical benefits achieved by eculizumab treatment of aHUS were maintained at 2 years of follow-up.
Collapse
|
35
|
Walle J, Delmas Y, Ardissino G, Wang J, Kincaid J, Haller H. Early initiation of eculizumab treatment in patients with atypical haemolytic uraemic syndrome improves long-term outcomes: a pooled analysis of clinical trials. Crit Care 2015. [PMCID: PMC4470673 DOI: 10.1186/cc14410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
36
|
Kambhampati S, Park W, Habtezion A. Pharmacologic therapy for acute pancreatitis. World J Gastroenterol 2014; 20:16868-16880. [PMID: 25493000 PMCID: PMC4258556 DOI: 10.3748/wjg.v20.i45.16868] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 07/23/2014] [Accepted: 10/15/2014] [Indexed: 02/06/2023] Open
Abstract
While conservative management such as fluid, bowel rest, and antibiotics is the mainstay of current acute pancreatitis management, there is a lot of promise in pharmacologic therapies that target various aspects of the pathogenesis of pancreatitis. Extensive review of preclinical studies, which include assessment of therapies such as anti-secretory agents, protease inhibitors, anti-inflammatory agents, and anti-oxidants are discussed. Many of these studies have shown therapeutic benefit and improved survival in experimental models. Based on available preclinical studies, we discuss potential novel targeted pharmacologic approaches that may offer promise in the treatment of acute pancreatitis. To date a variety of clinical studies have assessed the translational potential of animal model effective experimental therapies and have shown either failure or mixed results in human studies. Despite these discouraging clinical studies, there is a great clinical need and there exist several preclinical effective therapies that await investigation in patients. Better understanding of acute pancreatitis pathophysiology and lessons learned from past clinical studies are likely to offer a great foundation upon which to expand future therapies in acute pancreatitis.
Collapse
|
37
|
[Diagnostic and therapeutic guidelines of thrombotic microangiopathies of the Spanish Apheresis Group]. Med Clin (Barc) 2014; 144:331.e1-331.e13. [PMID: 25433791 DOI: 10.1016/j.medcli.2014.09.013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2014] [Accepted: 09/18/2014] [Indexed: 12/18/2022]
Abstract
Thrombotic microangiopathies (TMA) are disorders defined by the presence of a microangiopathic hemolytic anemia (with the characteristic hallmark of schistocytes in the peripheral blood smear), thrombocytopenia and organ malfunction of variable intensity. Thrombotic thrombocytopenic purpura and hemolytic uremic syndrome are the most important forms of TMA and, without the adequate treatment, they are associated with high morbimortality. In recent years, significant advances in the knowledge of the pathophysiology of TMA have occurred. Those advances have allowed us to move from a syndromic diagnosis with a similar treatment to all entities to the search of etiologic diagnosis which would lead to a specific treatment, finally leading to a better outcome of the patient. This document pretends to summarize the current status of knowledge of the pathophysiology of TMA and the therapeutic options available, and to offer a diagnostic and therapeutic practical tool to the professionals caring for the patients.
Collapse
|
38
|
Sharp JA, Whitley PH, Cunnion KM, Krishna NK. Peptide inhibitor of complement c1, a novel suppressor of classical pathway activation: mechanistic studies and clinical potential. Front Immunol 2014; 5:406. [PMID: 25202312 PMCID: PMC4141160 DOI: 10.3389/fimmu.2014.00406] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 08/08/2014] [Indexed: 11/20/2022] Open
Abstract
The classical pathway of complement plays multiple physiological roles including modulating immunological effectors initiated by adaptive immune responses and an essential homeostatic role in the clearance of damaged self-antigens. However, dysregulated classical pathway activation is associated with antibody-initiated, inflammatory diseases processes like cold agglutinin disease, acute intravascular hemolytic transfusion reaction (AIHTR), and acute/hyperacute transplantation rejection. To date, only one putative classical pathway inhibitor, C1 esterase inhibitor (C1-INH), is currently commercially available and its only approved indication is for replacement treatment in hereditary angioedema, which is predominantly a kinin pathway disease. Given the variety of disease conditions in which the classical pathway is implicated, development of therapeutics that specifically inhibits complement initiation represents a major unmet medical need. Our laboratory has identified a peptide that specifically inhibits the classical and lectin pathways of complement. In vitro studies have demonstrated that these peptide inhibitors of complement C1 (PIC1) bind to the collagen-like region of the initiator molecule of the classical pathway, C1q. PIC1 binding to C1q blocks activation of the associated serine proteases (C1s–C1r–C1r–C1s) and subsequent downstream complement activation. Rational design optimization of PIC1 has resulted in the generation of a highly potent derivative of 15 amino acids. PIC1 inhibits classical pathway mediated complement activation in ABO incompatibility in vitro and inhibiting classical pathway activation in vivo in rats. This review will focus on the pre-clinical development of PIC1 and discuss its potential as a therapeutic in antibody-mediated classical pathway disease, specifically AIHTR.
Collapse
Affiliation(s)
- Julia A Sharp
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School , Norfolk, VA , USA
| | | | - Kenji M Cunnion
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School , Norfolk, VA , USA ; Department of Pediatrics, Eastern Virginia Medical School , Norfolk, VA , USA ; Children's Specialty Group, Division of Infectious Diseases , Norfolk, VA , USA
| | - Neel K Krishna
- Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School , Norfolk, VA , USA ; Department of Pediatrics, Eastern Virginia Medical School , Norfolk, VA , USA
| |
Collapse
|
39
|
Qiao Q, Teng X, Wang N, Lu R, Guo L, Zhang X, Du Y, Wang W, Chen S, Wu Q, He G, Wang Y, Hu W. A novel CRIg-targeted complement inhibitor protects cells from complement damage. FASEB J 2014; 28:4986-99. [PMID: 25114177 DOI: 10.1096/fj.14-258046] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The inappropriate activation of complement may contribute to various immune diseases. The alternative pathway (AP) predominates during complement activation regardless of the initiating pathways. Hence, the main AP regulator factor H (FH) holds great potential as an attractive therapeutic intervention. In addition, complement receptor of the immunoglobulin superfamily (CRIg) has been demonstrated to inhibit AP and, more notably, still specifically binds to C3b/iC3b. We thus developed novel CRIg-targeted complement inhibitors by connecting the functional domains of CRIg and FH, which we termed CRIg-FH and CRIg-L-FH. CRIg-L-FH, slightly more potent than CRIg-FH, considerably inhibited both AP- and also classical pathway (CP)-mediated hemolysis and successfully eliminated the deposition of C3b/iC3b. Kinetic analysis further revealed that the binding affinity constant (KD) of CRIg/FH was in the micromolar range, consistent with its long-lasting binding to complement-attacked cells. CRIg-L-FH efficiently protected aberrant erythrocytes of patients with paroxysmal nocturnal hemoglobinuria (PNH) from AP- and CP-mediated complement damage (IC50 was 22.43 and 64.69 nM, respectively). Moreover, CRIg-L-FH was found to inhibit complement activation induced by the anti-Thy1 antibody in a mesangioproliferative glomerulonephritis (MPGN) rat model. Hence, CRIg-L-FH protects glomerular mesangial cells (GMCs) from complement-mediated injury and proliferative lesions. These findings strongly suggest that CRIg/FH is a potential therapeutic drug candidate for a range of complement-mediated diseases.
Collapse
Affiliation(s)
- Qian Qiao
- Shanghai Cancer Center, Institutes of Biomedical Sciences, Department of Oncology, and
| | - Xiaoyan Teng
- Shanghai Cancer Center, Institutes of Biomedical Sciences, Department of Oncology, and
| | - Na Wang
- Shanghai Cancer Center, Institutes of Biomedical Sciences, Department of Oncology, and
| | - Renquan Lu
- Shanghai Cancer Center, Institutes of Biomedical Sciences, Department of Oncology, and
| | - Lin Guo
- Shanghai Cancer Center, Institutes of Biomedical Sciences, Department of Oncology, and
| | - Xin Zhang
- Shanghai Cancer Center, Institutes of Biomedical Sciences, Department of Oncology, and
| | - Yiqun Du
- Shanghai Cancer Center, Institutes of Biomedical Sciences, Department of Oncology, and
| | - Wenjuan Wang
- Ministry of Health Key Laboratory of Thrombosis and Hematostasis, Jiangsu Institute of Hematology, Department of Hematology, First Affiliated Hospital of Soochow University, Suzhou, China; and
| | - Suning Chen
- Ministry of Health Key Laboratory of Thrombosis and Hematostasis, Jiangsu Institute of Hematology, Department of Hematology, First Affiliated Hospital of Soochow University, Suzhou, China; and
| | - Qian Wu
- Ministry of Health Key Laboratory of Thrombosis and Hematostasis, Jiangsu Institute of Hematology, Department of Hematology, First Affiliated Hospital of Soochow University, Suzhou, China; and
| | - Guangsheng He
- Ministry of Health Key Laboratory of Thrombosis and Hematostasis, Jiangsu Institute of Hematology, Department of Hematology, First Affiliated Hospital of Soochow University, Suzhou, China; and
| | - Yingwei Wang
- Department of Microbiology and Immunology, Nanjing Medical University, Nanjing, China
| | - Weiguo Hu
- Shanghai Cancer Center, Institutes of Biomedical Sciences, Department of Oncology, and Department of Immunology, Shanghai Medical College, Fudan University, Shanghai, China;
| |
Collapse
|
40
|
TNT003, an inhibitor of the serine protease C1s, prevents complement activation induced by cold agglutinins. Blood 2014; 123:4015-22. [DOI: 10.1182/blood-2014-02-556027] [Citation(s) in RCA: 106] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Key Points
Cold agglutinin–driven classical pathway activity terminates prior to the initiation of the terminal cascade in CAD patient blood. By inhibiting cold agglutinin–mediated complement deposition on the cellular membrane, TNT003 prevents RBCs from being phagocytosed.
Collapse
|
41
|
|