pubmed_id
stringlengths
41
43
abstract
stringlengths
3
18.8k
http://www.ncbi.nlm.nih.gov/pubmed/20041827
1. Curr Pharm Des. 2010;16(8):988-1001. doi: 10.2174/138161210790883480. Splice modification to restore functional dystrophin synthesis in Duchenne muscular dystrophy. Wilton SD(1), Fletcher S. Author information: (1)Centre for Neuromuscular and Neurological Disorders, University of Western Australia, 4th Floor A Block, QE II Medical Centre, Nedlands, Western Australia, Australia. [email protected] In little more than a decade, induced exon skipping as a therapy to treat Duchenne muscular dystrophy (DMD) has progressed from a concept tested in vitro, to pre-clinical evaluation in mouse and dog models, and recent completion of Phase I clinical trials in man. There is no longer any doubt that antisense oligomers can redirect dystrophin gene processing and by-pass protein truncating mutations after direct injection into muscle. Proof-of-concept has been demonstrated in human dystrophic muscle, with trials in Leiden and London showing that two different oligomer chemistries can restore the reading-frame in selected DMD patients by excising dystrophin exon 51. Systemic delivery of both oligomer types into DMD patients has commenced with promising results but it remains to be established if this therapy will have measurable clinical benefits. Targeted removal of exon 51 will only be directly applicable to about one in ten DMD individuals, and the immediate challenges include development of appropriate and effective delivery regimens, and extending splice-switching therapies to other dystrophin gene lesions. The success of induced exon skipping has spawned a number of "fusion therapies", including vector-mediated dystrophin exon skipping and ex vivo viral delivery of splice-switching antisense molecules into myogenic stem cells, followed by implantation, which may address long term oligomer delivery issues. This review summarizes the pivotal events leading to the completion of the first proof-of-concept trials and speculates on some of the scientific, ethical, regulatory and commercial challenges facing targeted exon skipping for the treatment of DMD. DOI: 10.2174/138161210790883480 PMID: 20041827 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/23609411
1. Nat Rev Genet. 2013 Jun;14(6):373-8. doi: 10.1038/nrg3460. Epub 2013 Apr 23. Therapy for Duchenne muscular dystrophy: renewed optimism from genetic approaches. Fairclough RJ(1), Wood MJ, Davies KE. Author information: (1)Department of Physiology, Anatomy and Genetics, University of Oxford, Parks Road, Oxford OX1 3PT, UK. Duchenne muscular dystrophy (DMD) is a devastating progressive disease for which there is currently no effective treatment except palliative therapy. There are several promising genetic approaches, including viral delivery of the missing dystrophin gene, read-through of translation stop codons, exon skipping to restore the reading frame and increased expression of the compensatory utrophin gene. The lessons learned from these approaches will be applicable to many other disorders. DOI: 10.1038/nrg3460 PMID: 23609411 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/17217326
1. Annu Rev Med. 2007;58:75-88. doi: 10.1146/annurev.med.58.011706.144703. New approaches in the therapy of cardiomyopathy in muscular dystrophy. McNally EM(1). Author information: (1)Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA. [email protected] Cardiomyopathy is a frequent occurrence in muscular dystrophy, and heart disease in muscular dystrophy can contribute to both morbidity and mortality. A number of novel therapies are being developed for muscular dystrophy, and the efficacy of these therapies for heart disease is unknown. The most common X-linked recessive disease is Duchenne muscular dystrophy (DMD), which arises from defects in the dystrophin gene. Therapy specifically aimed at DMD is reviewed in the context of its projected effect on cardiomyopathy associated with DMD. Additionally, novel therapies are being pursued to treat specifically the cardiomyopathy of DMD. There is substantial genetic heterogeneity underlying the muscular dystrophies, and not all muscular dystrophy patients develop cardiomyopathy. A subset of muscular dystrophies may place patients at significantly greater risk of developing cardiomyopathy and cardiac rhythm disturbances. These disorders are discussed, highlighting recent studies and recommendations for therapy. DOI: 10.1146/annurev.med.58.011706.144703 PMID: 17217326 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/19527108
1. Expert Opin Biol Ther. 2009 Jul;9(7):849-66. doi: 10.1517/14712590903029164. Gene therapy for muscular dystrophy: current progress and future prospects. Trollet C(1), Athanasopoulos T, Popplewell L, Malerba A, Dickson G. Author information: (1)School of Biological Sciences, Royal Holloway-University of London, Egham, TW20 0EX, Surrey, UK. Muscular dystrophies refer to a group of inherited disorders characterized by progressive muscle weakness, wasting and degeneration. So far, there is no effective treatment but new gene-based therapies are currently being developed with particular noted advances in using conventional gene replacement strategies, RNA-based approaches, or cell-based gene therapy with a main focus on Duchenne muscular dystrophy (DMD). DMD is the most common and severe form of muscular dystrophy and current treatments are far from adequate. However, genetic and cell-based therapies, in particular exon skipping induced by antisense strategies, and corrective gene therapy via functionally engineered dystrophin genes hold great promise, with several clinical trials ongoing. Proof-of-concept of exon skipping has been obtained in animal models, and most recently in clinical trials; this approach represents a promising therapy for a subset of patients. In addition, gene-delivery-based strategies exist both for antisense-induced reading frame restoration, and for highly efficient delivery of functional dystrophin mini- and micro-genes to muscle fibres in vivo and muscle stem cells ex-vivo. In particular, AAV-based vectors show efficient systemic gene delivery to skeletal muscle directly in vivo, and lentivirus-based vectors show promise of combining ex vivo gene modification strategies with cell-mediated therapies. DOI: 10.1517/14712590903029164 PMID: 19527108 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/23387802
1. FEBS J. 2013 Sep;280(17):4263-80. doi: 10.1111/febs.12178. Epub 2013 Mar 4. Repair or replace? Exploiting novel gene and cell therapy strategies for muscular dystrophies. Benedetti S(1), Hoshiya H, Tedesco FS. Author information: (1)Department of Cell and Developmental Biology, University College London, UK. Muscular dystrophies are genetic disorders characterized by skeletal muscle wasting and weakness. Although there is no effective therapy, a number of experimental strategies have been developed over recent years and some of them are undergoing clinical investigation. In this review, we highlight recent developments and key challenges for strategies based upon gene replacement and gene/expression repair, including exon-skipping, vector-mediated gene therapy and cell therapy. Therapeutic strategies for different forms of muscular dystrophy are discussed, with an emphasis on Duchenne muscular dystrophy, given the severity and the relatively advanced status of clinical studies for this disease. © 2013 The Authors Journal compilation © 2013 FEBS. DOI: 10.1111/febs.12178 PMID: 23387802 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35939579
1. N Engl J Med. 2022 Jul 28;387(4):332-344. doi: 10.1056/NEJMoa2117872. Germline Mutations in CIDEB and Protection against Liver Disease. Verweij N(1), Haas ME(1), Nielsen JB(1), Sosina OA(1), Kim M(1), Akbari P(1), De T(1), Hindy G(1), Bovijn J(1), Persaud T(1), Miloscio L(1), Germino M(1), Panagis L(1), Watanabe K(1), Mbatchou J(1), Jones M(1), LeBlanc M(1), Balasubramanian S(1), Lammert C(1), Enhörning S(1), Melander O(1), Carey DJ(1), Still CD(1), Mirshahi T(1), Rader DJ(1), Parasoglou P(1), Walls JR(1), Overton JD(1), Reid JG(1), Economides A(1), Cantor MN(1), Zambrowicz B(1), Murphy AJ(1), Abecasis GR(1), Ferreira MAR(1), Smagris E(1), Gusarova V(1), Sleeman M(1), Yancopoulos GD(1), Marchini J(1), Kang HM(1), Karalis K(1), Shuldiner AR(1), Della Gatta G(1), Locke AE(1), Baras A(1), Lotta LA(1). Author information: (1)From the Regeneron Genetics Center (N.V., M.E.H., J.B.N., O.A.S., M.K., P.A., T.D., G.H., J.B., T.P., L.M., K.W., J. Mbatchou, M.J., M.L., S.B., J.D.O., J.G.R., A.E., M.N.C., G.R.A., M.A.R.F., J. Marchini, H.M.K., K.K., A.R.S., G.D.G., A.E.L., A.B., L.A.L.), Regeneron Pharmaceuticals (M.G., L.P., P.P., J.R.W., B.Z., A.J.M., E.S., V.G., M.S., G.D.Y.), Tarrytown, NY; Indiana University School of Medicine, Indianapolis (C.L.); the Department of Clinical Sciences Malmö, Lund University, and the Department of Emergency and Internal Medicine, Skåne University Hospital - both in Malmö, Sweden (S.E., O.M.); and the Department of Molecular and Functional Genomics, Geisinger Health System, Danville (D.J.C., C.D.S., T.M.), and the Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (D.J.R.) - both in Pennsylvania. Comment in Gastroenterology. 2023 Feb;164(2):304-305. doi: 10.1053/j.gastro.2022.09.045. Liver Int. 2022 Dec;42(12):2594-2596. doi: 10.1111/liv.15460. BACKGROUND: Exome sequencing in hundreds of thousands of persons may enable the identification of rare protein-coding genetic variants associated with protection from human diseases like liver cirrhosis, providing a strategy for the discovery of new therapeutic targets. METHODS: We performed a multistage exome sequencing and genetic association analysis to identify genes in which rare protein-coding variants were associated with liver phenotypes. We conducted in vitro experiments to further characterize associations. RESULTS: The multistage analysis involved 542,904 persons with available data on liver aminotransferase levels, 24,944 patients with various types of liver disease, and 490,636 controls without liver disease. We found that rare coding variants in APOB, ABCB4, SLC30A10, and TM6SF2 were associated with increased aminotransferase levels and an increased risk of liver disease. We also found that variants in CIDEB, which encodes a structural protein found in hepatic lipid droplets, had a protective effect. The burden of rare predicted loss-of-function variants plus missense variants in CIDEB (combined carrier frequency, 0.7%) was associated with decreased alanine aminotransferase levels (beta per allele, -1.24 U per liter; 95% confidence interval [CI], -1.66 to -0.83; P = 4.8×10-9) and with 33% lower odds of liver disease of any cause (odds ratio per allele, 0.67; 95% CI, 0.57 to 0.79; P = 9.9×10-7). Rare coding variants in CIDEB were associated with a decreased risk of liver disease across different underlying causes and different degrees of severity, including cirrhosis of any cause (odds ratio per allele, 0.50; 95% CI, 0.36 to 0.70). Among 3599 patients who had undergone bariatric surgery, rare coding variants in CIDEB were associated with a decreased nonalcoholic fatty liver disease activity score (beta per allele in score units, -0.98; 95% CI, -1.54 to -0.41 [scores range from 0 to 8, with higher scores indicating more severe disease]). In human hepatoma cell lines challenged with oleate, CIDEB small interfering RNA knockdown prevented the buildup of large lipid droplets. CONCLUSIONS: Rare germline mutations in CIDEB conferred substantial protection from liver disease. (Funded by Regeneron Pharmaceuticals.). Copyright © 2022 Massachusetts Medical Society. DOI: 10.1056/NEJMoa2117872 PMID: 35939579 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34605036
1. Med Res Rev. 2022 Mar;42(2):654-709. doi: 10.1002/med.21856. Epub 2021 Oct 4. Analyzing the scaffold diversity of cyclin-dependent kinase inhibitors and revisiting the clinical and preclinical pipeline. Bhurta D(1)(2), Bharate SB(1)(2). Author information: (1)Natural Products & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu, India. (2)Academy of Scientific & Innovative Research (AcSIR), Ghaziabad, India. Kinases have gained an important place in the list of vital therapeutic targets because of their overwhelming clinical success in the last two decades. Among various clinically validated kinases, the cyclin-dependent kinases (CDK) are one of the extensively studied drug targets for clinical development. Food and Drug Administration has approved three CDK inhibitors for therapeutic use, and at least 27 inhibitors are under active clinical development. In the last decade, research and development in this area took a rapid pace, and thus the analysis of scaffold diversity is essential for future drug design. Available reviews lack the systematic study and discussion on the scaffold diversity of CDK inhibitors. Herein we have reviewed and critically analyzed the chemical diversity present in the preclinical and clinical pipeline of CDK inhibitors. Our analysis has shown that although several scaffolds represent CDK inhibitors, only the amino-pyrimidine is a well-represented scaffold. The three-nitrogen framework of amino-pyrimidine is a fundamental hinge-binding unit. Further, we have discussed the selectivity aspects among CDKs, the clinical trial dose-limiting toxicities, and highlighted the most advanced clinical candidates. We also discuss the changing paradigm towards selective inhibitors and an overview of ATP-binding pockets of all druggable CDKs. We carefully analyzed the clinical pipeline to unravel the candidates that are currently under active clinical development. In addition to the plenty of dual CDK4/6 inhibitors, there are many selective CDK7, CDK9, and CDK8/19 inhibitors in the clinical pipeline. © 2021 Wiley Periodicals LLC. DOI: 10.1002/med.21856 PMID: 34605036 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35628286
1. Int J Mol Sci. 2022 May 13;23(10):5476. doi: 10.3390/ijms23105476. Antitumoral Activity of a CDK9 PROTAC Compound in HER2-Positive Breast Cancer. Noblejas-López MDM(1)(2), Gandullo-Sánchez L(3), Galán-Moya EM(1)(2)(4), López-Rosa R(1)(2), Tébar-García D(1)(2), Nieto-Jiménez C(5), Gómez-Juárez M(1), Burgos M(1)(2)(6), Pandiella A(3), Ocaña A(1)(2)(5). Author information: (1)Translational Research Unit, Albacete University Hospital, 02008 Albacete, Spain. (2)Centro Regional de Investigaciones Biomédicas (CRIB), Castilla-La Mancha University (UCLM), 02008 Albacete, Spain. (3)Instituto de Biología Molecular y Celular del Cáncer, CSIC, IBSAL and CIBERONC, 37007 Salamanca, Spain. (4)Faculty of Nursing, Castilla-La Mancha University (UCLM), 02008 Albacete, Spain. (5)Experimental Therapeutics Unit, Medical Oncology Department, Hospital Clínico San Carlos (HCSC), Instituto de Investigación Sanitaria (IdISSC) and CIBERONC, 28040 Madrid, Spain. (6)Department of Nutrition, Food Science and Physiology, School of Pharmacy and Nutrition, University of Navarra, 31008 Pamplona, Spain. Cyclin-dependent kinases (CDKs) are a broad family of proteins involved in the cell cycle and transcriptional regulation. In this article, we explore the antitumoral activity of a novel proteolysis-targeting chimera (PROTAC) compound against CDK9. Breast cancer cell lines from different subtypes were used. Transcriptomic mapping of CDKs in breast cancer demonstrated that the expression of CDK9 predicted a detrimental outcome in basal-like tumors (HR = 1.51, CI = 1.08-2.11, p = 0.015) and, particularly, in the luminal B subtype with HER2+ expression (HR = 1.82, CI = 1.17-2.82, p = 0.0069). The novel CDK9 PROTAC, THAL-SNS-032, displayed a profound inhibitory activity in MCF7, T47D, and BT474 cells, with less effect in SKBR3, HCC1569, HCC1954, MDA-MB-231, HS578T, and BT549 cells. The three cell lines with HER2 overexpression and no presence of ER, SKBR3, HCC1569, and HCC1954 displayed an EC50 three times higher compared to ER-positive and dual ER/HER2-positive cell lines. BT474-derived trastuzumab-resistant cell lines displayed a particular sensitivity to THAL-SNS-032. Western blot analyses showed that THAL-SNS-032 caused a decrease in CDK9 levels in BT474, BT474-RH, and BT474-TDM1R cells, and a significant increase in apoptosis. Experiments in animals demonstrated an inverse therapeutic index of THAL-SNS-032, with doses in the nontherapeutic and toxic range. The identified toxicity was mainly due to an on-target off-tumor effect of the compound in the gastrointestinal epithelium. In summary, the potent and efficient antitumoral properties of the CDK9 PROTAC THAL-SNS-032 opens the possibility of using this type of compound in breast cancer only if specifically delivered to cancer cells, particularly in ER/HER2-positive and HER2-resistant tumors. DOI: 10.3390/ijms23105476 PMCID: PMC9146359 PMID: 35628286 [Indexed for MEDLINE] Conflict of interest statement: A.O. is currently an employee of Symphogen, Copengahen, Denmark.
http://www.ncbi.nlm.nih.gov/pubmed/31603123
1. J Cancer Res Ther. 2019 Jul-Sep;15(5):1131-1140. doi: 10.4103/jcrt.JCRT_47_18. Ligand- and structure-based pharmacophore modeling, docking study reveals 2-[[4-[6-(isopropylamino) pyrimidin-4-yl]-1H-pyrrolo[2,3-b] pyridin-6-yl] amino] ethanol as a potential anticancer agent of CDK9/cyclin T1 kinase. Hussain A(1), Verma CK(1). Author information: (1)Department of Bioinformatics, MANIT, Bhopal, Madhya Pradesh, India. OBJECTIVE: CDK9/Cyclin T1 kinase is a protein kinase, indirectly involved in the cell cycle progression in the form of transcription elongation, CDK9 specific inhibitors may be a potential alternative treatment not only for cancer but also other life-threatening diseases. MATERIALS AND METHODS: Ligand-based and structure-based pharmacophore model was developed for discovering of the new anticancer agents. These models used as three-dimensional query for virtual screening against the chemical structure databases such as Maybridge HitFinder, MDPI, and ZINC. Subsequently, the potential hit compound was filtered by the ADMET and docking score. RESULTS: After applying all filtration, 11 hits were found as potential hits based on good docking scores as well as good ADMET properties. Compound 2-[4-[6-(isopropylamino) pyrimidin-4-yl]-1H-pyrrolo[2,3-b] pyridin-6-yl] amino] ethanol was found to be most potent among all the potential hits. These hits could be used as an anticancer agent in near future. CONCLUSIONS: So many advances in the treatment of death leading diseases have been made over the past few decades, However, looking for the development in this research ligand-based and structure-based pharmacophore modeling was done, hit1 2-[4-[6-(isopropylamino) pyrimidin-4-yl]-1H-pyrrolo[2,3 b] pyridin-6 yl] amino] ethanol was found to be more potent and selective. It is understandable that these hits could be as selective and potent anticancer agents of cyclin-dependent kinase complex. DOI: 10.4103/jcrt.JCRT_47_18 PMID: 31603123 [Indexed for MEDLINE] Conflict of interest statement: None
http://www.ncbi.nlm.nih.gov/pubmed/12432243
1. Cancer Biol Ther. 2002 Jul-Aug;1(4):342-7. CDK9: from basal transcription to cancer and AIDS. De Falco G(1), Giordano A. Author information: (1)Istituto di Anatomia ed Istologia Patologica, Università degli Studi di Siena, Italy. Cdk9 is a member of the Cdc2-like family of kinases. Its cyclin partners are members of the family of cyclin T (T1, T2a and T2b) and cyclin K. The Cdk9/cyclin T complexes appear to be involved in regulating several physiological processes. Cdk9/cyclin T1 belongs to the P-TEFb complex, and is responsible for the phosphorylation of the carboxyl-terminal domain (CTD) of the RNA Polymerase II, thus promoting general elongation. Cdk9 has also been described as the kinase of the TAK complex, which is homologous to the P-TEFb complex and involved in HIV replication. Cdk9 also appears to be involved in the differentiation program of several cell types, such as muscle cells, monocytes and neurons, suggesting that it may have a function in controlling specific differentiative pathways. In addition, Cdk9 seems to have an anti-apoptotic function in monocytes, that may be related to its control over differentiation of monocytes. This data suggests the involvement of Cdk9 in several physiological processes in the cell, the deregulation of which may be related to the genesis of transforming events, that may in turn lead to the onset of cancer. In addition, since the complex Cdk9/cyclin T1 is able to bind to the HIV-1 product Tat, the study of the functions of Cdk9/cyclin T may be of interest in understanding the basal mechanisms that regulate HIV replication. PMID: 12432243 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/20150322
1. Brain. 2010 Apr;133(Pt 4):957-72. doi: 10.1093/brain/awq002. Epub 2010 Feb 11. RNA-targeted splice-correction therapy for neuromuscular disease. Wood MJ(1), Gait MJ, Yin H. Author information: (1)Department of Physiology, Anatomy and Genetics, University of Oxford, South Parks Road, Oxford OX1 3QX, UK. [email protected] Splice-modulation therapy, whereby molecular manipulation of premessenger RNA splicing is engineered to yield genetic correction, is a promising novel therapy for genetic diseases of muscle and nerve-the prototypical example being Duchenne muscular dystrophy. Duchenne muscular dystrophy is the most common childhood genetic disease, affecting one in 3500 newborn boys, causing progressive muscle weakness, heart and respiratory failure and premature death. No cure exists for this disease and a number of promising new molecular therapies are being intensively studied. Duchenne muscular dystrophy arises due to mutations that disrupt the open-reading-frame in the DMD gene leading to the absence of the essential muscle protein dystrophin. Of all novel molecular interventions currently being investigated for Duchenne muscular dystrophy, perhaps the most promising method aiming to restore dystrophin expression to diseased cells is known as 'exon skipping' or splice-modulation, whereby antisense oligonucleotides eliminate the deleterious effects of DMD mutations by modulating dystrophin pre-messenger RNA splicing, such that functional dystrophin protein is produced. Recently this method was shown to be promising and safe in clinical trials both in The Netherlands and the UK. These trials studied direct antisense oligonucleotide injections into single peripheral lower limb muscles, whereas a viable therapy will need antisense oligonucleotides to be delivered systemically to all muscles, most critically to the heart, and ultimately to all other affected tissues including brain. There has also been considerable progress in understanding how such splice-correction methods could be applied to the treatment of related neuromuscular diseases, including spinal muscular atrophy and myotonic dystrophy, where defects of splicing or alternative splicing are closely related to the disease mechanism. DOI: 10.1093/brain/awq002 PMID: 20150322 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/9766517
1. J Cell Physiol. 1998 Nov;177(2):206-13. doi: 10.1002/(SICI)1097-4652(199811)177:2<206::AID-JCP2>3.0.CO;2-R. Cloning of murine CDK9/PITALRE and its tissue-specific expression in development. Bagella L(1), MacLachlan TK, Buono RJ, Pisano MM, Giordano A, De Luca A. Author information: (1)Department of Pathology, Anatomy and Cell Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA. The cdc2-family of serine/threonine kinases and their binding partners recently were implicated in developmental roles. We previously cloned a cdc2-related kinase, cdk9/PITALRE, that is able to phosphorylate the retinoblastoma protein in vitro. We describe here the cloning and the characterization of the mouse homolog of cdk9/PITALRE. The murine cDNA is 98% identical with humans and is expressed at high levels in brain and kidney tissues. The kinase activity and protein expression of cdk9/PITALRE were highest in terminally differentiated tissues such as the muscle and brain. In situ immunohistology and immunofluorescence detected cdk9/PITALRE protein not only within terminally differentiated cells such as muscle and neuronal cells, but also in proliferating cells. C2C12 and P19 cells induced to differentiate along muscle and neural lineages peaked in cdk9/PITALRE kinase activity at the end of differentiation. These results suggest that, among other roles, cdk9/PITALRE plays a role not unlike cdk5 in the differentiation of certain cell types. DOI: 10.1002/(SICI)1097-4652(199811)177:2<206::AID-JCP2>3.0.CO;2-R PMID: 9766517 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/27833949
1. Mol Biosyst. 2017 Jan 31;13(2):246-276. doi: 10.1039/c6mb00387g. The emerging picture of CDK9/P-TEFb: more than 20 years of advances since PITALRE. Paparidis NF(1), Durvale MC(2), Canduri F(1). Author information: (1)Department of Chemistry and Molecular Physics, Institute of Chemistry of Sao Carlos, Sao Paulo University, Av. Trabalhador Sãocarlense, 400, Zip Code 780, 13560-970, São Carlos-SP, Brazil. [email protected]. (2)Department of Biochemistry, Institute of Chemistry, Sao Paulo University, Av. Prof. Lineu Prestes, 748, 05508-000, Butantã - São Paulo - SP, Brazil. CDK9 is a prominent member of the transcriptional CDKs subfamily, a group of kinases whose function is to control the primary steps of mRNA synthesis and processing by eukaryotic RNA polymerase II. As a cyclin-dependent kinase, CDK9 activation in vivo depends upon its association with T-type cyclins to assemble the positive transcription elongation factor (P-TEFb). Although CDK9/P-TEFb phosphorylates the C-terminal domain of RNAP II in the same positions targeted by CDK7 (TFIIH) and CDK8 (Mediator), the former does not participate in the transcription initiation, but rather plays a unique role by driving the polymerase to productive elongation. In addition to RNAP II CTD, the negative transcription elongation factors DSIF and NELF also represent major CDK9 substrates, whose phosphorylation is required to overcome the proximal pause of the polymerase. CDK9 is recruited to specific genes through proteins that interact with both P-TEFb and distinct elements in DNA, RNA or chromatin, where it modulates the activity of individual RNAP II transcription complexes. The regulation of CDK9 function is an intricate network that includes post-translational modifications (phosphorylation/dephosphorylation and acetylation/deacetylation of key residues) as well as the association of P-TEFb with various proteins that can stimulate or inhibit its kinase activity. Several cases of CDK9 deregulation have been linked to important human diseases, including various types of cancer and also AIDS (due to its essential role in HIV replication). Not only HIV, but also many other human viruses have been shown to depend strongly on CDK9 activity to be transcribed within host cells. This review summarizes the main advances made on CDK9/P-TEFb field in more than 20 years, introducing the structural, functional and genetic aspects that have been elucidated ever since. DOI: 10.1039/c6mb00387g PMID: 27833949 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/36454480
1. Cell Mol Life Sci. 2022 Dec 1;79(12):613. doi: 10.1007/s00018-022-04624-4. RBM24 controls cardiac QT interval through CaMKIIδ splicing. Liu J(#)(1)(2)(3), Wang K(#)(1), Liu X(4), Pan L(1), Zhou W(1), Huang J(1), Liu H(1)(2), Su Z(1)(2), Xu XQ(5)(6). Author information: (1)The Institute of Stem Cell and Regenerative Medicine, School of Medicine, Xiamen University, Xiamen, 361100, Fujian, People's Republic of China. (2)Department of Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361100, People's Republic of China. (3)Shenzhen Research Institute of Xiamen University, Shenzhen, 518000, Guangdong, People's Republic of China. (4)School of Medicine, Xiamen University, Xiamen, 361100, Fujian, People's Republic of China. (5)The Institute of Stem Cell and Regenerative Medicine, School of Medicine, Xiamen University, Xiamen, 361100, Fujian, People's Republic of China. [email protected]. (6)Department of Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, 361100, People's Republic of China. [email protected]. (#)Contributed equally Calcium/calmodulin-dependent kinase II delta (CaMKIIδ) is the predominant cardiac isoform and it is alternatively spliced to generate multiple variants. Variable variants allow for distinct localization and potentially different functions in the heart. Dysregulation of CaMKIIδ splicing has been demonstrated to be involved in the pathogenesis of heart diseases, such as cardiac hypertrophy, arrhythmia, and diastolic dysfunction. However, the mechanisms that regulate CaMKIIδ are incompletely understood. Here, we show that RNA binding motif protein 24 (RBM24) is a key splicing regulator of CaMKIIδ. RBM24 ablation leads to the aberrant shift of CaMKIIδ towards the δ-C isoform, which is known to activate the L-type Ca current. In line with this, we found marked alteration in Ca2+ handling followed by prolongation of the ventricular cardiac action potential and QT interval in RBM24 knockout mice, and these changes could be attenuated by treatment with an inhibitor of CaMKIIδ. Importantly, knockdown of RBM24 in human embryonic stem cell-derived cardiomyocytes showed similar electrophysiological abnormalities, suggesting the important role of RBM24 in the human heart. Thus, our data suggest that RBM24 is a critical regulator of CaMKIIδ to control the cardiac QT interval, highlighting the key role of splicing regulation in cardiac rhythm. © 2022. The Author(s), under exclusive licence to Springer Nature Switzerland AG. DOI: 10.1007/s00018-022-04624-4 PMID: 36454480 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35501714
1. BMC Neurol. 2022 May 2;22(1):162. doi: 10.1186/s12883-022-02687-1. Characteristics of disease progression and genetic correlation in ambulatory Iranian boys with Duchenne muscular dystrophy. Zamani G(#)(1), Hosseinpour S(#)(2), Ashrafi MR(1), Mohammadi M(1), Badv RS(1), Tavasoli AR(1), Akbari MG(3), Bereshneh AH(4), Malamiri RA(5), Heidari M(6). Author information: (1)Pediatrics Center of Excellence, Department of Pediatric Neurology, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran. (2)Department of Pediatric Neurology, Vali-e-Asr Hospital, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran. (3)Physical Medicine and Rehabilitation Department, Children's Medical Center , Tehran University of Medical Sciences, Tehran, Iran. (4)Prenatal Diagnosis and Genetic Research Center, Dastgheib Hospital, Shiraz University of Medical Sciences, Shiraz, Iran. [email protected]. (5)Department of Pediatric Neurology, Golestan Medical, Educational, and Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. (6)Pediatrics Center of Excellence, Department of Pediatric Neurology, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran. [email protected]. (#)Contributed equally BACKGROUND: Duchenne muscular dystrophy (DMD) is the most common muscular dystrophy in the pediatric population. The manifestations of this disease include progressive muscle weakness, gait dysfunction, and motor impairment, leading to a loss of ambulation by the age of 13 years. Molecular diagnosis is the standard diagnostic tool for DMD. This study aimed to investigate disease progression and genetic patterns in Iranian ambulant boys and to find the correlation between genotypes and motor function phenotypes. METHODS: This study was performed on 152 DMD patients. Clinical history, including the disease phenotype, steroid therapy, and the North Star Ambulatory Assessment (NSAA) score, was taken for all the patients. Molecular diagnoses were confirmed by multiplex ligation-dependent probe amplification and next-generation sequencing tests. RESULTS: A total of 152 Iranian DMD patients were examined in this study. The mean age at the time of disease onset was 4.04 ± 2.00 years, and the mean age at diagnosis was 5.05 ± 2.08 years. The mean age of ambulation loss was 10.9 years. Contracture was reported in 38.9% of cases. In terms of age, the mean total NSAA score showed a peak at 4 years of age, with a mean NSAA score of 24. Annual changes in the NSAA score were determined for all cases, based on the mutation type and exon site. Deletion mutation was found in 79.1% of cases, duplication in 6.8%, nonsense in 12.8%, and splice site in 1.4%. The most common single exon deletion was exon 44 (5.3%), and the most common multiexon deletions were attributed to exons 45-50 and exons 45-52 (4.6%). The results did not indicate any correlation between the mutation type and age at the time of disease onset, loss of ambulation age, and wheelchair dependence; however, a significant association was found between contracture and mutation type. The results showed a significant difference in the NSAA score between the deletion and nonsense groups at the age of 3 years (P = 0.04). No significant correlation was found between the phenotype and exon site. Overall, 91.1% of the study population had a history of corticosteroid use, and 54.1% showed compliance with rehabilitation therapy. CONCLUSION: This study demonstrated the phenotypes and mutational features of Iranian DMD boys and provided information regarding the natural motor history of the disease, disease progression, diagnosis, and status of DMD management in Iran. The present findings can promote the development of clinical trials and future advanced molecular therapies in Iran. © 2022. The Author(s). DOI: 10.1186/s12883-022-02687-1 PMCID: PMC9059913 PMID: 35501714 [Indexed for MEDLINE] Conflict of interest statement: We declare no financial conflicts of interest.
http://www.ncbi.nlm.nih.gov/pubmed/35626905
1. Children (Basel). 2022 May 16;9(5):728. doi: 10.3390/children9050728. Evaluating the Feasibility and Reliability of Remotely Delivering and Scoring the North Star Ambulatory Assessment in Ambulant Patients with Duchenne Muscular Dystrophy. Emery N(1), Strachan K(1), Kulshrestha R(1), Kuiper JH(1)(2), Willis T(1). Author information: (1)Neuromuscular Service, TORCH Building, Robert Jones and Agnes Hunt Orthopaedic Hopsital, Shropshire SY10 7AG, UK. (2)School of Pharmacy and Bioengineering, Keele University, Staffordshire ST5 5BG, UK. OBJECTIVE: The North Star Ambulatory Assessment (NSAA) is a validated 17-item functional rating scale and widely used to assess motor function in boys with Duchenne muscular dystrophy (DMD). The SARS-CoV-2 pandemic and subsequent Government 'lockdown' resulted in no face-to-face clinic visits hence the motor abilities were not monitored. The aim was to investigate whether the NSAA was feasible and reliable by video assessment. METHOD: Ten ambulant DMD boys were selected from the electronic hospital records. Two physiotherapists scored the boys' NSAA independently and the intraclass correlation coefficient was used to assess agreement. The video scores were compared to two previous NSAA in-clinic scores. RESULTS: Mean scores (SD) for clinic visit one were 22.6 (4.19) and clinic visit two 21.8 (5.3). The two physiotherapists video mean scores were 20.6 (5.66) for physiotherapist 1 and 20.6 (6.53) for physiotherapist 2. The intraclass correlation coefficient was 0.98 (95% CI 0.93-1.00) for the total NSAA and 1.00 (95% CI 1.00 to 1.00) for the rise time. The mean decline in score from clinic visit one (-12 months) to video assessment was 2.0 (2.8SD). CONCLUSION: The results from the study suggest that video NSAA is partially feasible and reliable. DOI: 10.3390/children9050728 PMCID: PMC9139300 PMID: 35626905 Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/34146121
1. Cell Mol Life Sci. 2021 Jul;78(14):5543-5567. doi: 10.1007/s00018-021-03878-8. Epub 2021 Jun 19. CDK9 keeps RNA polymerase II on track. Egloff S(1). Author information: (1)Molecular, Cellular and Developmental Biology Department (MCD), Centre de Biologie Intégrative (CBI), University of Toulouse, CNRS, UPS, 31062, Toulouse, France. [email protected]. Cyclin-dependent kinase 9 (CDK9), the kinase component of positive transcription elongation factor b (P-TEFb), is essential for transcription of most protein-coding genes by RNA polymerase II (RNAPII). By releasing promoter-proximally paused RNAPII into gene bodies, CDK9 controls the entry of RNAPII into productive elongation and is, therefore, critical for efficient synthesis of full-length messenger (m)RNAs. In recent years, new players involved in P-TEFb-dependent processes have been identified and an important function of CDK9 in coordinating elongation with transcription initiation and termination has been unveiled. As the regulatory functions of CDK9 in gene expression continue to expand, a number of human pathologies, including cancers, have been associated with aberrant CDK9 activity, underscoring the need to properly regulate CDK9. Here, I provide an overview of CDK9 function and regulation, with an emphasis on CDK9 dysregulation in human diseases. DOI: 10.1007/s00018-021-03878-8 PMCID: PMC8257543 PMID: 34146121 [Indexed for MEDLINE] Conflict of interest statement: The author declares no competing interests.
http://www.ncbi.nlm.nih.gov/pubmed/34062779
1. Cancers (Basel). 2021 May 1;13(9):2181. doi: 10.3390/cancers13092181. Targeting CDK9 for Anti-Cancer Therapeutics. Mandal R(1), Becker S(1), Strebhardt K(1)(2). Author information: (1)Department of Gynecology and Obstetrics, Johann Wolfgang Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany. (2)German Cancer Consortium (DKTK), 69120 Heidelberg, Germany. Cyclin Dependent Kinase 9 (CDK9) is one of the most important transcription regulatory members of the CDK family. In conjunction with its main cyclin partner-Cyclin T1, it forms the Positive Transcription Elongation Factor b (P-TEFb) whose primary function in eukaryotic cells is to mediate the positive transcription elongation of nascent mRNA strands, by phosphorylating the S2 residues of the YSPTSPS tandem repeats at the C-terminus domain (CTD) of RNA Polymerase II (RNAP II). To aid in this process, P-TEFb also simultaneously phosphorylates and inactivates a number of negative transcription regulators like 5,6-dichloro-1-β-D-ribofuranosylbenzimidazole (DRB) Sensitivity-Inducing Factor (DSIF) and Negative Elongation Factor (NELF). Significantly enhanced activity of CDK9 is observed in multiple cancer types, which is universally associated with significantly shortened Overall Survival (OS) of the patients. In these cancer types, CDK9 regulates a plethora of cellular functions including proliferation, survival, cell cycle regulation, DNA damage repair and metastasis. Due to the extremely critical role of CDK9 in cancer cells, inhibiting its functions has been the subject of intense research, resulting the development of multiple, increasingly specific small-molecule inhibitors, some of which are presently in clinical trials. The search for newer generation CDK9 inhibitors with higher specificity and lower potential toxicities and suitable combination therapies continues. In fact, the Phase I clinical trials of the latest, highly specific CDK9 inhibitor BAY1251152, against different solid tumors have shown good anti-tumor and on-target activities and pharmacokinetics, combined with manageable safety profile while the phase I and II clinical trials of another inhibitor AT-7519 have been undertaken or are undergoing. To enhance the effectiveness and target diversity and reduce potential drug-resistance, the future of CDK9 inhibition would likely involve combining CDK9 inhibitors with inhibitors like those against BRD4, SEC, MYC, MCL-1 and HSP90. DOI: 10.3390/cancers13092181 PMCID: PMC8124690 PMID: 34062779 Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/35862363
1. Dev Med Child Neurol. 2022 Dec;64(12):1453-1461. doi: 10.1111/dmcn.15345. Epub 2022 Jul 21. Quality of instruments assessing activity and participation of people with muscular dystrophy: A systematic review of participant-reported outcome measures. Andrade KKS(1), Soares LA(1), Macedo CC(1), Bispo NR(1), Sousa Junior RR(2), Oliveira VC(1), Leite HR(1)(2), Gaiad TP(1). Author information: (1)Department of Physical Therapy, Faculty of Health Sciences, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, Minas Gerais, Brazil. (2)School of Physical Education, Physiotherapy and Occupational Therapy, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil. AIM: To identify the standardized assessment scales for people with muscular dystrophy and investigate the quality/level of evidence of their measurement properties. METHOD: A systematic review of patient-reported outcome measures was conducted on the MEDLINE, Embase, AMED, DiTA, and PsycINFO databases in August 2020. We included psychometric studies that investigated the validity, reliability, and responsiveness of instruments assessing activity and participation for muscular dystrophy of any type (Duchenne, Becker, limb-girdle, facioscapulohumeral, congenital, and myotonic) or age. Two independent reviewers selected the studies, extracted data, and evaluated the instruments' quality and level of evidence following the COnsensus-based Standards for the selection of health status Measurement INstruments (COSMIN) checklist. The study followed the Preferred Reporting Items for Systematic reviews and Meta-Analysis (PRISMA) 2020 guidelines. RESULTS: The searches identified 6675 references; a total of 46 studies with 28 condition-specific or general instruments were included. The measurement properties of most instruments had sufficient (68.8%) or indeterminate (25.7%) results according to COSMIN. The quality of evidence of the measurement properties was moderate (23.8%) or low (22.6%) according to the Grading of Recommendations Assessment, Development, and Evaluation (GRADE). INTERPRETATION: There is a lack of high-quality instruments whose psychometric properties are adequately measured. The highest quality instrument is the Muscular Dystrophy Functional Rating Scale. The Motor Function Measure (general instrument), Duchenne Muscular Dystrophy Upper-limb Patient-reported Outcome Measure, North Star Ambulatory Assessment, and Myotonic Dystrophy Type 1 Activity and Participation Scale for Clinical Use (specific) are also recommended. WHAT THIS PAPER ADDS: There are 28 available instruments for activity and participation of people with muscular dystrophy. The evidence quality is moderate or low because of imprecision and indirectness. The Muscular Dystrophy Functional Rating Scale is the highest quality instrument. The Motor Function Measure is the second most recommended instrument. The Duchenne Muscular Dystrophy Upper-limb Patient-reported Outcome Measure, North Star Ambulatory Assessment, and Myotonic Dystrophy Type 1 Activity and Participation Scale for Clinical Use are also recommended. © 2022 Mac Keith Press. DOI: 10.1111/dmcn.15345 PMID: 35862363 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/21200140
1. Cell Cycle. 2011 Jan 1;10(1):28-32. doi: 10.4161/cc.10.1.14364. Epub 2011 Jan 1. A role for CDK9-cyclin K in maintaining genome integrity. Yu DS(1), Cortez D. Author information: (1)Department of Radiation Oncology, Emory University School of Medicine, Atlanta, GA, USA. [email protected] Cyclin-dependent kinase 9 (CDK9), with its cyclin T regulatory subunit, is a component of the positive transcription elongation factor b (P-TEFb) complex, which stimulates transcription elongation and also functions in co-transcriptional histone modification, mRNA processing, and mRNA export. CDK9 also binds to cyclin K but the function of this CDK9-cyclin K complex is less clear. We and others have recently shown that CDK9 functions directly in maintaining genome integrity. This activity is restricted to CDK9-cyclin K. Depletion of CDK9 or its cyclin K but not cyclin T regulatory subunit impairs cell cycle recovery in response to replication stress and induces spontaneous DNA damage in replicating cells. CDK9-cyclin K also interacts with ATR and other DNA damage response and DNA repair proteins. CDK9 accumulates on chromatin and limits the amount of single-stranded DNA in response to replication stress. Collectively, these data are consistent with a model in which CDK9 responds to replication stress by localizing to chromatin to reduce the breakdown of stalled replication forks and promote recovery from replication arrest. The direct role of CDK9-cyclin K in pathways that maintain genome integrity in response to replication stress appear to be evolutionarily conserved. DOI: 10.4161/cc.10.1.14364 PMCID: PMC3048070 PMID: 21200140 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35618576
1. Neuromuscul Disord. 2022 Jun;32(6):460-467. doi: 10.1016/j.nmd.2022.02.012. Epub 2022 Feb 26. Functional outcome measures in young, steroid-naïve boys with Duchenne muscular dystrophy. Mayhew AG(1), Moat D(2), McDermott MP(3), Eagle M(4), Griggs RC(5), King WM(6), James MK(2), Muni-Lofra R(2), Shillington A(7), Gregson S(7), Pallant L(8), Skura C(9), Staudt LA(9), Eichinger K(5), McMurchie H(10), Rabb R(10), Di Marco M(11), Brown S(11), Zanin R(12), Arnoldi MT(12), McIntyre M(13), Wilson A(13), Alfano LN(14), Lowes LP(14), Blomgren C(15), Milev E(16), Iodice M(16), Pasternak A(17), Chiu A(18), Lehnert I(19), Claus N(19), Dieruf KA(20), Rolle E(21), Nicorici A(22), Andres B(23), Hobbiebrunken E(24), Roetmann G(24), Kern V(25), Civitello M(26), Vogt S(27), Hayes MJ(28), Scholtes C(29), Lacroix C(30), Gunn T(31), Warner S(31), Newman J(32), Barp A(33), Kundrat K(34), Kovelman S(34), Powers PJ(35), Guglieri M(2); Muscle Study Group and TREAT-NMD. Author information: (1)John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University, International Centre for Life, Central Parkway, Newcastle Upon Tyne, NE1 3BZ, United Kingdom. Electronic address: [email protected]. (2)John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University, International Centre for Life, Central Parkway, Newcastle Upon Tyne, NE1 3BZ, United Kingdom. (3)Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States; Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, NY, United States. (4)John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University, International Centre for Life, Central Parkway, Newcastle Upon Tyne, NE1 3BZ, United Kingdom; ATOM International Limited, Gateshead, United Kingdom. (5)Department of Neurology, University of Rochester Medical Center, Rochester, NY, United States. (6)Department of Neurology, Neuromuscular Disease Center, The Ohio State University, OH, United States. Electronic address: [email protected]. (7)Alder Hey Children's NHS Foundation Trust, Liverpool, United Kingdom. (8)Leeds General Infirmary, Leeds Children's Hospital, United Kingdom. (9)Neurology Department, David Geffen School of Medicine at UCLA, Los Angeles, California, United States. (10)Heartlands Hospital, University Hospitals Birmingham, Birmingham, United Kingdom. (11)Royal Hospital for Children, Glasgow, United Kingdom. (12)IRCCS Istituto Neurologico Carlo Besta, Milan, Italy. (13)Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, UT, United States. (14)The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, United States; Department of Pediatrics, The Ohio State University, Columbus, OH, United States. (15)Department of Rehabilitation Services, Ann and Robert H Lurie Children's Hospital of Chicago, IL, United States. (16)Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom. (17)Department of Physical and Occupational Therapy Services, Boston Children's Hospital, Boston, MA, United States. (18)Neuroscience Program, Alberta Children's Hospital, Calgary, Canada. (19)Department of Neuropediatrics, Medical Faculty Carl Gustav Carus, Technical University Dresden, Germany. (20)University of New Mexico, Albuquerque, NM, United States. (21)Neuromuscular Unit, Department of Neurosciences, University of Torino, Italy. (22)Neuromuscular Research Lab in the Department of PM&R at UC Davis Health, Sacramento, CA, United States. (23)University Hospital Essen, Germany. (24)Department of Pediatrics and Adolescent Medicine, Division of Pediatric Neurology, University Medical Center Göttingen, Germany. (25)Penn State Health Hershey Medical Center, Hershey, PA, United States. (26)Nemours Children's Hospital, Orlando, FL, United States; St Jude Children's Research Hospital, Memphis, TN, United States. (27)Department of Neurosciences, University Children Hospital of Freiburg, Germany. (28)Department of Neurology, University of Kansas Medical Center, Kansas, United States. (29)London Health Science Centre, London ON, Canada. (30)Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada. (31)Royal Manchester Children's Hospital, Manchester, United Kingdom. (32)UNC Health Care - Rehabilitation Services, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States. (33)Department of Neurosciences, University of Padova, Padua, Italy. (34)Children's National Hospital, Washington, DC, United States. (35)Vanderbilt University Medical Center, Phi Beta Phi Rehabilitation Institute, Nashville, TN, United States. The purpose of this study was to quantitate motor performance in 196 genetically confirmed steroid-naïve boys with Duchenne muscular dystrophy (DMD), to evaluate the test-retest reliability of measures of motor performance in young DMD boys, and to assess correlations among the different functional outcomes including timed tests. Boys aged 4-7 years were recruited in the FOR-DMD study, a comparative effectiveness study of different steroid regimens in DMD. Eligible boys had to be able to rise from the floor independently and to perform pulmonary function testing consistently. The boys were evaluated with standardized assessments at the screening and baseline visits at 32 sites in 5 countries (US, UK, Canada, Italy, Germany). Assessments included timed rise from floor, timed 10 m walk/run, six-minute walk distance, North Star Ambulatory Assessment (NSAA) and forced vital capacity (FVC). Mean age at baseline was 5.9 years (range 4.1-8.1 years). Test-retest reliability was high for functional assessments, regardless of time lag between assessments (up to 90 days) and for the majority of age groups. Correlations were strong among the functional measures and timed tests, less so with FVC. Physiotherapy measures are reliable in a young, steroid-naïve population and rise from floor velocity appears to be a sensitive measure of strength in this population. Copyright © 2022 The Authors. Published by Elsevier B.V. All rights reserved. DOI: 10.1016/j.nmd.2022.02.012 PMID: 35618576 [Indexed for MEDLINE] Conflict of interest statement: Declaration of Competing Interests Anna G. Mayhew has participated in SAB meetings for Roche, Regenxbio, Summit, PTC and Biogen and performs Consultancy work (training physiotherapists for trial in DMD) for Roche, Pfizer, PTC, Summit, Sarepta, Lysogene, Italfarmaco, Amicus, Biogen and Avexis. Dionne Moat provides consultancy services for the following companies: ATOM International (covers consultancy services provided to Amicus Therapeutics Pty Ltd, Ascendis Pharma, Biomarin, Catabasis, Faraday, FibroGen, Genethon, Italfarmaco, NS Pharma, Pfizer, PTC Therapeutics, QED Therapeutics Ltd, Reveragen, Sarepta Therapeutics), outside the submitted work. Michael McDermott has been supported by a research grant from PTC Therapeutics, has received compensation for consulting from Fulcrum Therapeutics, Inc., and NeuroDerm, Ltd., and has served on Data and Safety Monitoring Boards (DSMBs) for AstraZeneca, Eli Lilly and Company, Catabasis Pharmaceuticals, Inc., Vaccinex, Inc., Cynapsus Therapeutics, Neurocrine Biosciences, Inc., Voyager Therapeutics, Prilenia Therapeutics Development, Ltd., ReveraGen BioPharma, Inc., and NS Pharma, Inc. Michelle Eagle is Managing Director at ATOM International Limited and is contracted with the following companies through ATOM: Amicus, Biomarin, Capricor, Catabasis, Denali, Dyne, Edgewise, Faraday, Fibrogen, Genethon, Italfarmaco, Lysogen, Modis, NSPharma, PTC, Sarepta, Solid, Referagen. Robert Griggs receives research support from the NIH, the Muscular Dystrophy Association, the Parent Project for Muscular Dystrophy, and from Sarepta Biopharma, PTC Therapeutics and Santhera Pharmaceuticals. He serves as a Consultant for Strongbridge and Stealth Pharmaceuticals. He is Chair of a DSMB for Solid Pharmaceuticals. He receives monies for editorial work from Elsevier as an Editor of Cecil Essentials and Cecil Textbook of Medicine. He serves as Chair of the Research Advisory Committee and is a Board Member of the American Brain Foundation. He is on the Executive Committee of the Muscle Study Group, which receives support for its activities from Pharmaceutical companies. Meredith K. James provides consultancy services for the following companies: ATOM International (covers consultancy services provided to Amicus Therapeutics Pty Ltd, Ascendis Pharma, Biomarin, Catabasis, Faraday, FibroGen, Genethon, Italfarmaco, NS Pharma, Pfizer, PTC Therapeutics, QED Therapeutics Ltd, Reveragen, Sarepta Therapeutics), outside the submitted work. MKJ has received payment for participation on advisory boards for F. Hoffman La Roche AG, PTC Therapeutics and fee support for PhD studies from the Jain Foundation, outside the submitted work. Robert Muni-Lofra has participated in the advisory board for Biogen and Roche and performs consultancy work (training physiotherapists for trial in DMD and SMA) for Roche, Pfizer, PTC, Summit, Sarepta, Italfarmaco, Amicus, Biogen and Avexis. Lindsey Pallant has performed consultancy work (workshops and training physiotherapists in trials for DMD) for PTC and Sarepta. Katy Eichinger has received personal compensation for serving on advisory boards and/or as a consultant for Ionis, Biogen, Acceleron, Fulcrum, Avidity, PTC and the Myotonic Dystrophy Foundation. Dr. Eichinger has received personal compensation for serving as a speaker from Cure SMA, FSH Society, and Ology. She has received research/grant support from the CMTA. Heather McMurchie has received financial support from PTC and Biogen to access and provide training. Marina DiMarco attended a World Muscle Society conference sponsored by PTC Therapeutics and is a Consultant Physiotherapist presenting at PTC workshops. Lindsay N. Alfano provides consultancy services through ATOM International, Ltd for the following companies: Amicus Therapeutics Pty Ltd, Catabasis, Genethon, Italfarmaco, NS Pharma, Pfizer, PTC Therapeutics); reports royalties and other support from Sarepta Therapeutics; royalties for licensed technologies; other support from Novartis Gene Therapies; advisory board for Biogen. Linda P. Lowes provides consultancy services through ATOM International, Ltd for the following companies: Amicus Therapeutics Pty Ltd, Catabasis, Genethon, Italfarmaco, NS Pharma, Pfizer, PTC Therapeutics); reports royalties and other support from Sarepta Therapeutics; royalties for licensed technologies; other support from Novartis Gene Therapies; advisory board for Biogen. Amy Pasternak has participated in advisory boards for Roche, Biogen, Avexis, Scholar Rock and Audentes. Victoria Kern has received an honorarium from the MDA USA for a presentation. Cheryl Scholtes has sat on advisor panels for Biogen Canada and Roche Canada, and has been provided education on behalf of Biogen Canada and Roche Canada. Catherine Lacroix has acted as a consultant and advisory board participant to Biogen. Katherine Kundrat has participated in an advisory board for Biogen. Michela Guglieri is the study chair for a ReveraGen study. MG has research collaborations with ReveraGen and Sarepta. MG is (has been over the past 5 years) PI for clinical trials sponsored by Pfizer, Italfarmaco, Santhera, ReveraGen, Dynacure, Roche, PTC, Summit. MG has been participating in advisory boards for Pfizer, NS Pharma, Dyne (consultancies through Newcastle University). MG has performed consultancy work (speaker) for Sarepta. Declarations of interest: none for remaining authors
http://www.ncbi.nlm.nih.gov/pubmed/11532614
1. Front Biosci. 2001 Sep 1;6:D1073-82. doi: 10.2741/simone. New insight in cdk9 function: from Tat to MyoD. Simone C(1), Giordano A. Author information: (1)Dept. of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA. Cdk9 is a serine-threonine cdc2-related kinase and its activity is not cell cycle-regulated. Cdk9 function depends on its kinase activity and also on its regulatory units: the T-family cyclins and cyclin K. Recently, several studies confirmed the role of cdk9 in different cellular processes such as signal transduction, basal transcription, HIV-Tat- and MyoD-mediated transcription and differentiation. All the referred data strongly support the concept of a multifunctional protein kinase with specific cytoplasmic and nuclear functions. DOI: 10.2741/simone PMID: 11532614 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35998119
1. PLoS One. 2022 Aug 23;17(8):e0272858. doi: 10.1371/journal.pone.0272858. eCollection 2022. Novel approaches to analysis of the North Star Ambulatory Assessment (NSAA) in Duchenne muscular dystrophy (DMD): Observations from a phase 2 trial. Muntoni F(1), Guglieri M(2), Mah JK(3), Wagner KR(4), Brandsema JF(5), Butterfield RJ(6), McDonald CM(7), Mayhew AG(8), Palmer JP(9), Marraffino S(9), Charnas L(9), Mercuri E(10). Author information: (1)NIHR Great Ormond Street Hospital Biomedical Research Centre, Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, London, United Kingdom. (2)The John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom. (3)Cumming School of Medicine, University of Calgary, Alberta Children's Hospital, Calgary, Alberta, Canada. (4)Center for Genetic Muscle Disorders, and Departments of Neurology and Neuroscience, Kennedy Krieger Institute, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America. (5)Division of Neurology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America. (6)University of Utah School of Medicine, Salt Lake City, Utah, United States of America. (7)Lawson Health Research Institute, Children's Hospital, London, Ontario, Canada. (8)Institute of Genetic Medicine, Newcastle University, Newcastle, United Kingdom. (9)Pfizer Inc, Cambridge, Massachusetts, United States of America. (10)Paediatric Neurology, Catholic University, and Centro Clinico Nemo, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy. INTRODUCTION: The North Star Ambulatory Assessment (NSAA) tool is a key instrument for measuring clinical outcomes in patients with Duchenne muscular dystrophy (DMD). To gain a better understanding of the longitudinal utility of the NSAA, we evaluated NSAA data from a phase II trial of 120 patients with DMD treated with domagrozumab or placebo. METHODS: The NSAA exploratory analyses included assessment of individual skills gained/lost, total skills gained/lost, cumulative loss of function, and the impact of transient loss of function due to a temporary disability on NSAA total score (temporary zero score). RESULTS: There was no significant difference in the total number of NSAA skills gained (mean 1.41 and 1.04, respectively; p = 0.3314) or lost (3.90 vs. 5.0; p = 0.0998) between domagrozumab- vs. placebo-treated patients at week 49. However, domagrozumab-treated patients were less likely to lose the ability to perform a NSAA item (hazard ratio 0.80, 95% confidence interval [CI]: 0.65-0.98, p = 0.029) over 48-weeks vs. placebo-treated patients. When temporary zero scores were changed to "not obtainable" (8 values from 7 patients), domagrozumab-treated patients scored higher on the NSAA total score versus placebo-treated patients (difference at week 49: 2.0, 95% CI: 0.1-3.9, p = 0.0359). CONCLUSIONS: These exploratory analyses reveal additional approaches to interpreting the NSAA data beyond just change in NSAA total score. These observations also highlight the importance of reporting items as "not obtainable" for a patient with a temporary/transient physical disability that impacts their ability to perform the NSAA test. CLINICALTRIALS.GOV IDENTIFIER: NCT02310763. DOI: 10.1371/journal.pone.0272858 PMCID: PMC9397979 PMID: 35998119 [Indexed for MEDLINE] Conflict of interest statement: F Muntoni has received support from, and/or has served as a paid consultant for, Avexis, Biogen, Dyne Therapeutics, Roche, Novartis, Pfizer, PTC Therapeutics, Sarepta Therapeutics, Santhera, the National Institute of Health Research Biomedical Research Centre at Great Ormond Street Hospital for Children NHS Foundation Trust, and University College London. M Guglieri has received support from, and/or has served as a paid consultant for, PTC Therapeutic, Capricorn, Marathon Pharmaceutical, Pfizer, ReveraGen, Italfarmaco SpA, WAVE Life Sciences, Santhera, Roche, Sarepta and Dynacure; and grant funding from H2020 and NIH. J Mah has received support from Pfizer, Sarepta Therapeutics, Bristol-Myers Squibb, Roche, NS Pharma, ReveraGen Biopharma, Catabasis, Italfarmaco SpA, and PTC Pharmaceuticals. K Wagner has received support from, and/or has served as a paid consultant for, Dynacure, PTC Therapeutics, Roche, Asklepios Biopharmaceutical Inc, Pfizer, Sarepta Therapeutics, Catabasis, and Vita Therapeutics. K Wagner is currently an employee of F. Hoffmann-La Roche Ag. JF Brandsema has received support from, and/or has served as a paid consultant for, Alexion, Audentes, AveXis, Biogen, Genentech, PTC Therapeutics, Sarepta, WAVE Life Sciences, CSL Behring, Cytokinetics, Fibrogen, Pfizer, and Summit. RJ Butterfield has received support from, and/or has served as a paid consultant for, Avexis, Caspricor Therapudics, Catabasis, PTC Therapeutics, Sarepta Therapeutics, Pfizer, Biogen, Avexis, and Acceleron. CM McDonald has received support from, and/or has served as a paid consultant for, PTC Therapeutics, Santhera Pharmaceuticals, Catabasis Pharmaceuticals Inc, Sarepta Therapeutics, Prosensa, Biomarin Pharmaceutical, Pfizer, Eli Lilly, Halo Therapeutics, Marathon Pharmaceuticals, Bristol-Myers Squibb, Novartis, Italfarmaco, Astellas / Mitobridge, Epirium Bio (formerly Cardero Therapeutics), Edgewise Therapeutics, Avidity Biosciences, Entrada Therapeutics. AG Mayhew has received support from, and/or has served as a paid consultant for, PTC Therapeutics, Biogen, BMS, Roche, Sarepta, Santhera, Italfarmaco, Catabasis, Mallinckrodt, Pfizer, Summit, Wave Biogen, and Avexis. J Palmer, L Charnas, and S Marraffino are employees of and own stock in Pfizer. EM Mercuri has worked as a PI and on advisory boards for Pfizer, PTC Therapeutics, Roche, Sarepta Therapeutics, Santhera, and NS. This does not alter our adherence to PLOS ONE policies on sharing data and materials.
http://www.ncbi.nlm.nih.gov/pubmed/35385138
1. Dev Med Child Neurol. 2022 Aug;64(8):979-988. doi: 10.1111/dmcn.15176. Epub 2022 Feb 14. Peak functional ability and age at loss of ambulation in Duchenne muscular dystrophy. Zambon AA(1)(2), Ayyar Gupta V(1), Ridout D(3)(4), Manzur AY(1)(4), Baranello G(1), Trucco F(1)(5), Muntoni F(1)(4); UK Northstar Clinical Network. Collaborators: Tirupath S, Douglas M, McFetridge J, Parasuraman D, Alhaswani Z, McMurchie H, Rabb R, Majumdar A, Vijayakumar K, Amin S, Mason F, Frimpong-Ansah C, Gibbon F, Parson B, Naismith K, Burslem J, Baxter A, Eadie C, Horrocks I, Di Marco M, Childs AM, Pallant L, Spinty S, Shillington A, Gregson S, Cheshman L, Wraige E, Gowda V, Jungbluth H, Sheehan J, Hughes I, Warner S, Straub V, Guglieri M, Mayhew A, Chow G, Williamson S, Willis T, Kulshrestha R, Emery N, Ramdas S, Ramjattan H, de Goede C, Selley A, Ong M, White K, Illingworth M, Geary M, Palmer J, White C, Greenfield K, Hewawitharana G, Julien Y, Stephens E, Tewnion J, Ambegaonkar G, Krishnakumar D, Taylor J, Ward C, Willis T, Wright E, Rylance C. Author information: (1)Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, UK. (2)Neuromuscular Repair Unit, Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy. (3)Population, Policy and Practice Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, UK. (4)NIHR Great Ormond Street Hospital Biomedical Research Centre, London, UK. (5)Children's Sleep Medicine, Evelina Children Hospital - Paediatric Respiratory Department Royal Brompton Hospital, Guy's and St Thomas' Trust, London, UK. AIM: To correlate the North Star Ambulatory Assessment (NSAA) and timed rise from floor (TRF) recorded at age of expected peak with age at loss of ambulation (LOA) in Duchenne muscular dystrophy (DMD). METHOD: Male children with DMD enrolled in the UK North Start Network database were included according to the following criteria: follow-up longer than 3 years, one NSAA record between 6 years and 7 years 6 months (baseline), at least one visit when older than 8 years. Data about corticosteroid treatment, LOA, genotype, NSAA, and TRF were analysed. Age at LOA among the different groups based on NSAA and TRF was determined by log-rank tests. Cox proportional hazard models were used for multivariable analysis. RESULTS: A total of 293 patients from 13 different centres were included. Mean (SD) age at first and last visit was 5 years 6 months (1 year 2 months) and 12 years 8 months (2 years 11 months) (median follow-up 7 years 4 months). Higher NSAA and lower TRF at baseline were associated with older age at LOA (p<0.001). Patients scoring NSAA 32 to 34 had a probability of 0.61 of being ambulant when older than 13 years compared with 0.34 for those scoring 26 to 31. In multivariable analysis, NSAA, TRF, and corticosteroid daily regimen (vs intermittent) were all independently associated with outcome (p=0.01). INTERPRETATION: Higher functional abilities at peak are associated with older age at LOA in DMD. This information is important for counselling families. These baseline measures should also be considered when designing clinical trials. © 2022 The Authors. Developmental Medicine & Child Neurology published by John Wiley & Sons Ltd on behalf of Mac Keith Press. DOI: 10.1111/dmcn.15176 PMCID: PMC9303180 PMID: 35385138 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/28648683
1. Neuromuscul Disord. 2017 Aug;27(8):723-729. doi: 10.1016/j.nmd.2017.05.013. Epub 2017 May 26. Reliability and validity analyses of the North Star Ambulatory Assessment in Brazilian Portuguese. Okama LO(1), Zampieri LM(1), Ramos CL(1), Toledo FO(1), Alves CRJ(2), Mattiello-Sverzut AC(2), Mayhew A(3), Sobreira CFR(4). Author information: (1)Department of Neurosciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil. (2)Department of Biomechanics, Medicine and Rehabilitation of the Locomotor Apparatus, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil. (3)John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine, International Centre for Life, Newcastle Upon Tyne, United Kingdom. (4)Department of Neurosciences, Ribeirão Preto Medical School, University of São Paulo, São Paulo, Brazil. Electronic address: [email protected]. The North Star Ambulatory Assessment measures motor performance in ambulatory boys with Duchenne muscular dystrophy, a hereditary and degenerative muscle disorder. To use the North Star Ambulatory Assessment in Brazilian boys, we performed the cross-cultural adaptation to the Portuguese language spoken in Brazil and evaluated the reliability and validity of the instrument. Cross-cultural adaptation included: independent translations, synthesis, committee review, pre-testing in 12 boys, back-translation and comparison with the original instrument. Thirty-five boys with Duchenne muscular dystrophy and 38 healthy age-matched controls were recruited for further analyses. Reliability was assessed by internal consistency and reproducibility. Validity studies included face, content, construct and known-groups analyses. Cross-cultural adaptation resulted in an adequate instrument. Reliability studies demonstrated high internal consistency (Cronbach's alpha = 0.935) and adequate intra and inter-rater reproducibility (intraclass correlation coefficient = 0.988 and 0.962). Validation analyses indicated satisfactory content, face and convergent construct validities, with positive correlations with the Motor Function Measure total score (r = 0.863) and the 6-minute walk test (r = 0.433). The known group validity was demonstrated by higher scores in younger boys with Duchenne muscular dystrophy (p = 0.005). North Star Ambulatory Assessment in Brazilian Portuguese is a reliable and valid instrument to measure functional capacity in boys with Duchenne muscular dystrophy. Copyright © 2017 Elsevier B.V. All rights reserved. DOI: 10.1016/j.nmd.2017.05.013 PMID: 28648683 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31479456
1. PLoS One. 2019 Sep 3;14(9):e0221097. doi: 10.1371/journal.pone.0221097. eCollection 2019. Categorising trajectories and individual item changes of the North Star Ambulatory Assessment in patients with Duchenne muscular dystrophy. Muntoni F(1)(2), Domingos J(1), Manzur AY(1), Mayhew A(3), Guglieri M(3); UK NorthStar Network; Sajeev G(4)(5), Signorovitch J(4)(5), Ward SJ(4). Author information: (1)Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health & Great Ormond Street Hospital, London, United Kingdom. (2)National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, United Kingdom. (3)John Walton Muscular Dystrophy Research Centre, Newcastle University, Newcastle, United Kingdom. (4)Collaborative Trajectory Analysis Project, Cambridge, Massachusetts, United States of America. (5)Analysis Group Inc., Boston, Massachusetts, United States of America. Functional variability among boys with Duchenne muscular dystrophy (DMD) is well recognised and complicates interpretation of clinical studies. We hypothesised that boys with DMD could be clustered into groups sharing similar trajectories of ambulatory function over time, as measured by the North Star Ambulatory Assessment (NSAA) total score. We also explored associations with other variables such as age, functional abilities, and genotype. Using the NorthStar Clinical Network database, 395 patients with >1 NSAA assessment were identified. We utilised latent class trajectory analysis of longitudinal NSAA scores, which produced evidence for at least four clusters of boys sharing similar trajectories versus age in decreasing order of clinical severity: 25% of the boys were in cluster 1 (NSAA falling to ≤ 5 at age ~10y), 35% were in cluster 2 (NSAA ≤ 5 ~12y), 21% in were cluster 3 (NSAA≤ 5 ~14y), and 19% in cluster 4 (NSAA > 5 up to 15y). Mean ages at diagnosis of DMD were similar across clusters (4.2, 3.9, 4.3, and 4.8y, respectively). However, at the first NSAA assessment, a significant (p<0.05) association was observed between earlier declining clusters and younger age, worse NSAA, slower rise from supine, slower 10 metre walk/run times, and younger age of steroid initiation. In order to assess the probability of observing complete loss of function for individual NSAA items, we examined the proportion of patients who shifted from a score of 1 or 2 at baseline to a score of 0. We also assessed the probability of gain of function using the inverse assessment and stratified the probability of deterioration, improvement-or static behavior-by age ranges and using baseline functional status. Using this tool, our study provides a comprehensive assessment of the NSAA in a large population of patients with DMD and, for the first time, describes discrete clusters of disease progression; this will be invaluable for future DMD clinical trial design and interpretation of findings. DOI: 10.1371/journal.pone.0221097 PMCID: PMC6719875 PMID: 31479456 [Indexed for MEDLINE] Conflict of interest statement: I have read the journal's policy and the authors of this manuscript have the following competing interests: G.S. and J.S. are employees of Analysis Group Inc., which has received research funding from the study sponsors via the Collaborative Trajectory Analysis Project. S.J.W. is an independent consultant who has received funding from the study sponsors via cTAP, and has also received funds from patient foundations (CureDuchenne, Parent Project Muscular Dystrophy) to establish the Collaborative Trajectory Analysis Project This does not alter our adherence to PLOS ONE policies on sharing data and materials.
http://www.ncbi.nlm.nih.gov/pubmed/25454732
1. Neuromuscul Disord. 2015 Jan;25(1):14-8. doi: 10.1016/j.nmd.2014.09.015. Epub 2014 Oct 6. Suitability of North Star Ambulatory Assessment in young boys with Duchenne muscular dystrophy. De Sanctis R(1), Pane M(1), Sivo S(1), Ricotti V(2), Baranello G(3), Frosini S(4), Mazzone E(1), Bianco F(1), Fanelli L(1), Main M(2), Corlatti A(3), D'Amico A(5), Colia G(5), Scalise R(1), Palermo C(1), Alfonsi C(1), Tritto G(1), Romeo DM(1), Graziano A(1), Battini R(4), Morandi L(3), Bertini E(5), Muntoni F(2), Mercuri E(6). Author information: (1)Department of Paediatric Neurology, Catholic University, Rome, Italy. (2)Dubowitz Neuromuscular Centre, Institute of Child Health, University College, London, United Kingdom. (3)Developmental Neurology Unit, C. Besta Neurological Institute IRCCS, Milan, Italy. (4)Department of Developmental Neuroscience, IRCSS Stella Maris, Pisa, Italy. (5)Unit of Neuromuscular and Neurodegenerative diseases, Department of Neurosciences, Bambino Gesù Children's Hospital, Rome, Italy. (6)Department of Paediatric Neurology, Catholic University, Rome, Italy. Electronic address: [email protected]. The aim of this study was to establish the suitability of the North Star Ambulatory Assessment for use in young boys with Duchenne muscular dystrophy. We studied 147 typically developing and 144 boys affected by Duchenne muscular dystrophy between the ages of 3 and 5 years. More than 85% of the typically developing boys by the age of 4 years had full scores on all the items with total scores ≥33/34. Before the age of 4 years more than 15% of the typically developing boys did not achieve full scores on all the items. Some items, such as standing on one leg, showed significant improvement with age. In contrast, other activities were rarely achieved even in the older boys. Even if there was a progressive increase in scores with age, both total and individual item scores in Duchenne were still far from those obtained in the typically developing children of the same age. Our findings suggest that the North Star Ambulatory Assessment can be reliably used at least from the age of 4 years. Longitudinal natural history data studies are needed to assess possible changes over time and the possible effect of early steroids. Copyright © 2014 Elsevier B.V. All rights reserved. DOI: 10.1016/j.nmd.2014.09.015 PMID: 25454732 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/21410696
1. Dev Med Child Neurol. 2011 Jun;53(6):535-42. doi: 10.1111/j.1469-8749.2011.03939.x. Epub 2011 Mar 17. Moving towards meaningful measurement: Rasch analysis of the North Star Ambulatory Assessment in Duchenne muscular dystrophy. Mayhew A(1), Cano S, Scott E, Eagle M, Bushby K, Muntoni F; North Star Clinical Network for Paediatric Neuromuscular Disease. Collaborators: Manzur AY, Muntoni F, Robb S, Main M, Kemp J, Ricotti V, Scott E, Bushby K, Straub V, Sarkozy A, Strehle E, Venkateswaran R, Eagle E, Mayhew A, Roper H, McMurchie H, Grace A, Spinty S, Peachey G, Shillington S, Quinlivan R, Groves L, Wraige E, Jungbluth H, Sheehan J, Spahr R, Hughes I, Bateman E, Cammiss C, Childs AM, Pallant L, Psyden K, Baxter P, Naismith K, Keddie A, Horrocks I, McWilliam R, Di Marco M, Hartley L, Sheen B, Fenton-May J, Jardine P, Majumdar A, Jenkins L, Chow G, Miah A, de Goede C, Thomas N, Geary M, Keslake K, White C, Greenfield K, MacAuley S, Baxter A, Yirrell Y, Longman C. Author information: (1)Institute of Human Genetics, International Centre for Life, Newcastle University, Newcastle upon Tyne, UK. [email protected] AIM: Reliable measurement of disease progression and the effect of therapeutic interventions in Duchenne muscular dystrophy (DMD) require clinically meaningful and scientifically sound rating scales. Therefore, we need robust evidence to support such tools. The North Star Ambulatory Assessment (NSAA) is a promising, clinician-rated scale with potential uses spanning clinical practice and clinical trials. In this study, we used Rasch analysis to test its suitability in these roles as a measurement instrument. METHOD: NSAA data from 191 ambulant boys (mean age at assessment 7 y 8 mo, SD 2 y 4 mo; range 3 y 6 mo-15 y 5 mo) with a confirmed diagnosis of DMD were examined for psychometric properties including clinical meaning, targeting, response categories, model fit, reliability, dependency, stability, and raw to interval-level measurement. All analyses were performed using the Rasch Unidimensional Measurement Model. RESULTS: Overall, Rasch analysis supported the NSAA as being a reliable (high Person Separation Index of 0.91) and valid (good targeting, little misfit, no reversed thresholds) measure of ambulatory function in DMD. One item displayed misfit (lifts head, fit residual 6.9) and there was evidence for some local dependency (stand on right/left leg, climb and descend box step right/left leg, and hop on right/left leg, residual correlations >0.40), which we provide potential solutions for in future use of the NSAA. Importantly, our findings supported good clinical validity in that the hierarchy of items within the scale produced by the analyses was supported by clinical opinion, thus increasing the clinical interpretability of scale scores. INTERPRETATION: In general, Rasch analysis supported the NSAA as a psychometrically robust scale for use in DMD clinical research and trials. This study also demonstrates how Rasch analysis is a useful instrument to detect and understand the key measurement issues of rating scales. © The Authors. Developmental Medicine & Child Neurology © 2011 Mac Keith Press. DOI: 10.1111/j.1469-8749.2011.03939.x PMID: 21410696 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31629611
1. Neuromuscul Disord. 2019 Nov;29(11):857-862. doi: 10.1016/j.nmd.2019.09.010. Epub 2019 Sep 26. Longitudinal natural history in young boys with Duchenne muscular dystrophy. Coratti G(1), Brogna C(1), Norcia G(2), Ricotti V(3), Abbott L(3), D'Amico A(4), Berardinelli A(5), Vita GL(6), Lucibello S(1), Messina S(6), Sansone V(7), Albamonte E(7), Colia G(4), Salmin F(7), Gardani A(5), Manzur A(3), Main M(3), Baranello G(8), Arnoldi MT(9), Parsons J(10), Carry T(10), Connolly AM(11), Bertini E(4), Muntoni F(12), Pane M(2), Mercuri E(13). Author information: (1)Paediatric Neurology, Catholic University, Rome, Italy; Centro Clinico Nemo, Policlinico Gemelli, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy. (2)Centro Clinico Nemo, Policlinico Gemelli, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy. (3)Dubowitz Neuromuscular Centre University College London, London, United Kingdom. (4)Unit of Neuromuscular and Neurodegenerative Disorders, Department of Neurosciences, Bambino Gesù Children's Research Hospital IRCCS, Rome, Italy. (5)Child and Adolescent Unit, IRCCS Mondino Foundation, Pavia, Italy. (6)Department of Clinical and Experimental Medicine, University of Messina and Nemo Sud Clinical Centre, Messina, Italy. (7)The NEMO Center in Milan, Neurorehabilitation Unit, University of Milan, ASST Niguarda Hospital, Milan, Italy. (8)Dubowitz Neuromuscular Centre University College London, London, United Kingdom; UO Neurologia dello Sviluppo, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy; NIHR Great Ormond Street Hospital Biomedical Research Centre, London, United Kingdom. (9)UO Neurologia dello Sviluppo, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy. (10)Department of Pediatrics and Neurology, University of Colorado, Denver, CO, United States. (11)Nationwide Children's Hospital, Ohio State University, Columbus, Ohio, United States. (12)Dubowitz Neuromuscular Centre University College London, London, United Kingdom; NIHR Great Ormond Street Hospital Biomedical Research Centre, London, United Kingdom. (13)Paediatric Neurology, Catholic University, Rome, Italy; Centro Clinico Nemo, Policlinico Gemelli, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy. Electronic address: [email protected]. The aim of this prospective multicentric study was to document disease progression in young boys affected by Duchenne muscular dystrophy (DMD) between age 3 and 6 years (±3 months) using the North Star Ambulatory Assessment scale. One hundred fifty-three DMD boys (573 assessments) younger than 6 years (mean: 4.68, SD: 0.84) with a genetically proven DMD diagnoses were included. Our results showed North Star Ambulatory Assessment scores progressively increased with age. The largest increase was observed between age 3 and 4 years but further increase was steadily observed until age of 6 years. Using a multiple linear regression analysis, we found that both the use of corticosteroids and the site of mutation significantly contributed to the North Star Ambulatory Assessment changes (p < 0.001). At each age point, boys on corticosteroid treatment had higher scores than corticosteroid naïve ones (p < 0.001). Similarly, patients with mutations downstream exon 44, had lower baseline scores and lower magnitude of changes compared to those with mutations located at the 5' end of the gene (p < 0,001). Very few boys achieved the age appropriate maximum score. These results provide useful information for the assessment and counselling of young DMD boys and for the design of clinical trials in this age group. Copyright © 2019 Elsevier B.V. All rights reserved. DOI: 10.1016/j.nmd.2019.09.010 PMID: 31629611 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35302767
1. J Med Chem. 2022 Apr 14;65(7):5300-5316. doi: 10.1021/acs.jmedchem.1c01170. Epub 2022 Mar 18. Discovery of Reversible Covalent Bruton's Tyrosine Kinase Inhibitors PRN473 and PRN1008 (Rilzabrutinib). Owens TD(1), Brameld KA(1), Verner EJ(1), Ton T(1), Li X(1), Zhu J(1), Masjedizadeh MR(1), Bradshaw JM(1), Hill RJ(1), Tam D(1), Bisconte A(1), Kim EO(1), Francesco M(1), Xing Y(1), Shu J(1), Karr D(1), LaStant J(1), Finkle D(1), Loewenstein N(1), Haberstock-Debic H(1), Taylor MJ(1), Nunn P(1), Langrish CL(1), Goldstein DM(1). Author information: (1)Principia Biopharma, a Sanofi Company, 220 E Grand Ave, South San Francisco, California 94080, United States. Bruton's tyrosine kinase (BTK), a Tec family tyrosine kinase, is critical in immune pathways as an essential intracellular signaling element, participating in both adaptive and immune responses. Currently approved BTK inhibitors are irreversible covalent inhibitors and limited to oncology indications. Herein, we describe the design of covalent reversible BTK inhibitors and the discoveries of PRN473 (11) and rilzabrutinib (PRN1008, 12). These compounds have exhibited potent and durable inhibition of BTK, in vivo efficacy in rodent arthritis models, and clinical efficacy in canine pemphigus foliaceus. Compound 11 has completed phase 1 trials as a topical agent, and 12 is in phase 3 trials for pemphigus vulgaris and immune thrombocytopenia. DOI: 10.1021/acs.jmedchem.1c01170 PMID: 35302767 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35301810
1. Clin Transl Sci. 2022 Jun;15(6):1507-1518. doi: 10.1111/cts.13271. Epub 2022 Apr 3. A thorough QTc study to evaluate the effects of oral rilzabrutinib administered alone and with ritonavir in healthy subjects. Ucpinar S(1), Darpo B(2), Neale A(1), Nunn P(1), Shu J(1), Chu KA(1), Kavanagh M(1), Xue H(2), Phiasivongsa P(1), Thomas D(1), Smith PF(3). Author information: (1)Principia Biopharma Inc., A Sanofi Company, South San Francisco, California, USA. (2)ERT, Philadelphia, Pennsylvania, USA. (3)Certara, Princeton, New Jersey, USA. This study aimed to define the clinically relevant supratherapeutic dose of rilzabrutinib, an oral Bruton tyrosine kinase (BTK) inhibitor, and evaluate potential effects of therapeutic and supratherapeutic exposures on cardiac repolarization in healthy subjects. This was a two-part phase I study (anzctr.org.au ACTRN12618001036202). Part A was a randomized, open-label, three-period, single-dose crossover study (n = 12) with rilzabrutinib 100 mg ± ritonavir 100 mg or rilzabrutinib 1200 mg. Part B was a randomized, double-blind, placebo-controlled, four-way, single-dose crossover study (n = 39) with matched placebo, rilzabrutinib 400 mg ± ritonavir 100 mg, or moxifloxacin (positive control). Primary objectives: part A - pharmacokinetics (PK) of rilzabrutinib ± ritonavir, safety, and optimal dose for Part B; Part B - effect of rilzabrutinib therapeutic and supratherapeutic concentration on electrocardiogram (ECG) parameters. ECGs and PK samples were serially recorded before and post-dose. In part A, rilzabrutinib 100 mg + ritonavir led to 17-fold area under the concentration-time curve (AUC0-∞ ) and 7-fold maximum plasma concentration (Cmax ) increases over rilzabrutinib alone. Rilzabrutinib 1200 mg was discontinued due to mild-to-moderate gastrointestinal intolerance. In Part B, rilzabrutinib 400 mg + ritonavir increased rilzabrutinib mean AUC0-∞ from 454 to 3800 ng h/mL and Cmax from 144 to 712 ng/mL. The concentration-QTc relationship was slightly negative, shallow (-0.01 ms/ng/mL [90% CI -0.016 to -0.001]), and an effect >10 ms on QTcF could be excluded within the observed range of plasma concentrations, up to 2500 ng/mL. Safety was similar to other studies of rilzabrutinib. In conclusion, rilzabrutinib, even at supratherapeutic doses, had no clinically relevant effects on ECG parameters, including the QTc interval. © 2022 The Authors. Clinical and Translational Science published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics. DOI: 10.1111/cts.13271 PMCID: PMC9199881 PMID: 35301810 [Indexed for MEDLINE] Conflict of interest statement: S.U., A.N., P.N., J.S., K.A.C., M.K., and P.P are or were former employees of and received stock ownership from Principia Biopharma Inc., a Sanofi company at the time of the study. B.D. reports stock or other ownership with ERT and consulting/advisory role as his main employment (excluding Principia Biopharma Inc., a Sanofi company). H.X. is an employee of ERT. D.T. was a former employee of and received stock ownership from Principia Biopharma Inc., a Sanofi company at the time of the study, and reports pending patent regarding rilzabrutinib. P.F.S. reports employment with Certara and received consulting fees from Principia Biopharma Inc, a Sanofi company.
http://www.ncbi.nlm.nih.gov/pubmed/20634072
1. Neuromuscul Disord. 2010 Nov;20(11):712-6. doi: 10.1016/j.nmd.2010.06.014. Epub 2010 Jul 14. North Star Ambulatory Assessment, 6-minute walk test and timed items in ambulant boys with Duchenne muscular dystrophy. Mazzone E(1), Martinelli D, Berardinelli A, Messina S, D'Amico A, Vasco G, Main M, Doglio L, Politano L, Cavallaro F, Frosini S, Bello L, Carlesi A, Bonetti AM, Zucchini E, De Sanctis R, Scutifero M, Bianco F, Rossi F, Motta MC, Sacco A, Donati MA, Mongini T, Pini A, Battini R, Pegoraro E, Pane M, Pasquini E, Bruno C, Vita G, de Waure C, Bertini E, Mercuri E. Author information: (1)Department of Paediatric Neurology, Catholic University, Rome, Italy. The North Star Ambulatory Assessment is a functional scale specifically designed for ambulant boys affected by Duchenne muscular dystrophy (DMD). Recently the 6-minute walk test has also been used as an outcome measure in trials in DMD. The aim of our study was to assess a large cohort of ambulant boys affected by DMD using both North Star Assessment and 6-minute walk test. More specifically, we wished to establish the spectrum of findings for each measure and their correlation. This is a prospective multicentric study involving 10 centers. The cohort included 112 ambulant DMD boys of age ranging between 4.10 and 17 years (mean 8.18±2.3 DS). Ninety-one of the 112 were on steroids: 37/91 on intermittent and 54/91 on daily regimen. The scores on the North Star assessment ranged from 6/34 to 34/34. The distance on the 6-minute walk test ranged from 127 to 560.6 m. The time to walk 10 m was between 3 and 15 s. The time to rise from the floor ranged from 1 to 27.5 s. Some patients were unable to rise from the floor. As expected the results changed with age and were overall better in children treated with daily steroids. The North Star assessment had a moderate to good correlation with 6-minute walk test and with timed rising from floor but less with 10 m timed walk/run test. The 6-minute walk test in contrast had better correlation with 10 m timed walk/run test than with timed rising from floor. These findings suggest that a combination of these outcome measures can be effectively used in ambulant DMD boys and will provide information on different aspects of motor function, that may not be captured using a single measure. Copyright © 2010. Published by Elsevier B.V. DOI: 10.1016/j.nmd.2010.06.014 PMID: 20634072 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/36182620
1. Transfus Med Rev. 2022 Oct;36(4):175-180. doi: 10.1016/j.tmrv.2022.08.001. Epub 2022 Sep 6. New Therapies for the Treatment of Warm Autoimmune Hemolytic Anemia. Fattizzo B(1), Barcellini W(2). Author information: (1)Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy. Electronic address: [email protected]. (2)Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy. Electronic address: [email protected]. In this review article we provide a critical insight into recent reports evaluating innovative therapies for warm type autoimmune hemolytic anemia (wAIHA). Among published articles, we selected two reports on the use of the proteasome inhibitor bortezomib in association with dexamethasone or rituximab, one study on the spleen tyrosine kinase inhibitor fostamatinib, and a retrospective study on recombinant erythropoietin (rEPO). Among recent scientific communications, we discussed a report on the phosphoinositide 3-kinase delta inhibitor (PI3Kδi) parsaclisib. All studies highlighted a good efficacy although to be confirmed in larger trials and with limitations due to the heterogeneity of wAIHA patients enrolled, the small number of subjects, the concomitant medications allowed, and the short follow-up. Ongoing trials include new B-cell/plasma-cell targeting agents such as the Bruton tyrosine kinase inhibitors ibrutinib and rilzabrutinib, and the anti-CD38 MoAbs daratumumab and its analogue isatuximab. Further drugs in clinical trials target the complement cascade in wAIHA with complement activation, such as the C3 inhibitor pegcetacoplan and the C1q inhibitor ANX005. Finally, an interesting and non-immuno-toxic strategy is to remove the pathogenic autoantibodies via blocking the neonatal Fc receptor, by intravenous nipocalimab and subcutaneous RVT-1401. Such novel agents targeting the several immunopathological mechanisms acting in wAIHA and their possible combination, will increase the therapeutic armamentarium and possibly fill the gap of wAIHA relapsed after/refractory to rituximab. Moreover, these new target therapies may represent a tool for the unmet need of very acute cases. Copyright © 2022. Published by Elsevier Inc. DOI: 10.1016/j.tmrv.2022.08.001 PMID: 36182620 [Indexed for MEDLINE] Conflict of interest statement: Conflict of interest statement BF received consultancy for Amgen, Alexion, Annexon, Apellis, Momenta, Novartis, Sobi. WB received consultancy for Agios, Alexion, Annexon, Sanofi, Novartis, Sobi.
http://www.ncbi.nlm.nih.gov/pubmed/35562130
1. Indian J Pathol Microbiol. 2022 May;65(Supplement):S14-S23. doi: 10.4103/ijpm.ijpm_34_22. A review of adult-type diffuse gliomas in the WHO CNS5 classification with special reference to Astrocytoma, IDH-mutant and Oligodendroglioma, IDH-mutant and 1p/19q codeleted. Santosh V(1), Rao S(1). Author information: (1)Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India. The fifth edition of the World Health Organization (WHO) Classification of Tumors of the Central Nervous System (WHO CNS5) features several changes in the classification, diagnostic criteria, nomenclature, and grading of diffuse gliomas. Adult-type diffuse gliomas are genetically defined and include astrocytoma, isocitrate dehydrogenase (IDH)-mutant, oligodendroglioma, IDH-mutant and 1p/19q codeleted, and glioblastoma, IDH-wildtype. This review briefly discusses two tumor types: astrocytoma, IDH-mutant, and oligodendroglioma, IDH-mutant and 1p/19q codeleted, with emphasis on relevant changes in their classification and defining molecular genetic alterations. A simplified approach to the diagnosis of these tumors is provided. DOI: 10.4103/ijpm.ijpm_34_22 PMID: 35562130 [Indexed for MEDLINE] Conflict of interest statement: None
http://www.ncbi.nlm.nih.gov/pubmed/35587830
1. Eur Radiol. 2022 Nov;32(11):7780-7788. doi: 10.1007/s00330-022-08850-z. Epub 2022 May 19. Diffuse glioma, not otherwise specified: imaging-based risk stratification achieves histomolecular-level prognostication. Jang EB(1), Kim HS(2), Park JE(1), Park SY(3), Nam YK(4), Nam SJ(5), Kim YH(6), Kim JH(6). Author information: (1)Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea. (2)Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea. [email protected]. (3)Department of Statistics and Data Science, Korea National Open University, Seoul, Republic of Korea. (4)Department of Radiology, Shinchon Yonsei Hospital, Seoul, Republic of Korea. (5)Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea. (6)Department of Neurosurgery, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea. Comment in Eur Radiol. 2022 Nov;32(11):7778-7779. doi: 10.1007/s00330-022-09098-3. OBJECTIVES: To determine whether imaging-based risk stratification enables prognostication in diffuse glioma, NOS (not otherwise specified). METHODS: Data from 220 patients classified as diffuse glioma, NOS, between January 2011 and December 2020 were retrospectively included. Two neuroradiologists analyzed pre-surgical CT and MRI to assign gliomas to the three imaging-based risk types considering well-known imaging phenotypes (e.g., T2/FLAIR mismatch). According to the 2021 World Health Organization classification, the three risk types included (1) low-risk, expecting oligodendroglioma, isocitrate dehydrogenase (IDH)-mutant, and 1p/19q-codeleted; (2) intermediate-risk, expecting astrocytoma, IDH-mutant; and (3) high-risk, expecting glioblastoma, IDH-wildtype. Progression-free survival (PFS) and overall survival (OS) were estimated for each risk type. Time-dependent receiver operating characteristic analysis using 10-fold cross-validation with 100-fold bootstrapping was used to compare the performance of an imaging-based survival model with that of a historical molecular-based survival model published in 2015, created using The Cancer Genome Archive data. RESULTS: Prognostication according to the three imaging-based risk types was achieved for both PFS and OS (log-rank test, p < 0.001). The imaging-based survival model showed high prognostic value, with areas under the curves (AUCs) of 0.772 and 0.650 for 1-year PFS and OS, respectively, similar to the historical molecular-based survival model (AUC = 0.74 for PFS and 0.87 for OS). The imaging-based survival model achieved high long-term performance in both 3-year PFS (AUC = 0.806) and 5-year OS (AUC = 0.812). CONCLUSION: Imaging-based risk stratification achieved histomolecular-level prognostication in diffuse glioma, NOS, and could aid in guiding patient referral for insufficient or unsuccessful molecular diagnosis. KEY POINTS: • Three imaging-based risk types enable distinct prognostication in diffuse glioma, NOS (not otherwise specified). • The imaging-based survival model achieved similar prognostic performance as a historical molecular-based survival model. • For long-term prognostication of 3 and 5 years, the imaging-based survival model showed high performance. © 2022. The Author(s), under exclusive licence to European Society of Radiology. DOI: 10.1007/s00330-022-08850-z PMID: 35587830 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31175262
1. Pract Neurol. 2019 Oct;19(5):412-416. doi: 10.1136/practneurol-2018-002116. Epub 2019 Jun 6. Clinical importance of molecular markers of adult diffuse glioma. Molinari E(1), Curran OE(2), Grant R(3). Author information: (1)Department of Neurology, Queen Elizabeth University Hospital, Glasgow, UK. (2)Neuropathology Unit, Department of Pathology, Western General Hospital, Edinburgh, UK. (3)Department of Neurology and Neurosurgery, Western General Hospital, Edinburgh, UK [email protected]. In 2016, the WHO incorporated molecular markers, in addition to histology, into the diagnostic classification of central nervous system (CNS) tumours. This improves diagnostic accuracy and prognostication: oligo-astrocytoma no longer exists as a clinical entity; isocitrate dehydrogenase (IDH) mutant and 1p/19q co-deleted oligodendroglioma is a smaller category with better prognosis; IDH wild-type 'low-grade' glioma has a much poorer prognosis; and glioblastoma is divided into IDH mutant (with an better prognosis than pre-2016 glioblastoma) and IDH wild type (with a poorer prognosis). Previous advice based on phenotype alone will change with respect to median survival, best management plan and response to treatment. There are implications for routine neuropathology reporting and future trial design. Cases that are difficult to classify may need more advanced molecular genetic classification through DNA methylation-based classification of CNS tumours (Heidelberg Classifier). We discuss the practical implications. © Author(s) (or their employer(s)) 2019. No commercial re-use. See rights and permissions. Published by BMJ. DOI: 10.1136/practneurol-2018-002116 PMID: 31175262 [Indexed for MEDLINE] Conflict of interest statement: Competing interests: None declared.
http://www.ncbi.nlm.nih.gov/pubmed/36006639
1. JAMA Oncol. 2022 Oct 1;8(10):1493-1501. doi: 10.1001/jamaoncol.2022.2844. World Health Organization 2021 Classification of Central Nervous System Tumors and Implications for Therapy for Adult-Type Gliomas: A Review. Berger TR(1)(2), Wen PY(1)(2)(3), Lang-Orsini M(4), Chukwueke UN(1)(2)(3). Author information: (1)Division of Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. (2)Harvard Medical School, Boston, Massachusetts. (3)Division of Neuro-Oncology, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts. (4)Division of Neuropathology, Department of Pathology, Massachusetts General Hospital, Boston. IMPORTANCE: Previous histologic classifications of brain tumors have been limited by discrepancies in diagnoses reported by neuropathologists and variability in outcomes and response to therapies. Such diagnostic discrepancies have impaired clinicians' ability to select the most appropriate therapies for patients and have allowed heterogeneous populations of patients to be enrolled in clinical trials, hindering the development of more effective therapies. In adult-type diffuse gliomas, histologic classification has a particularly important effect on clinical care. OBSERVATIONS: In 2021, the World Health Organization published the fifth edition of the Classification of Tumors of the Central Nervous System. This classification incorporates advances in understanding the molecular pathogenesis of brain tumors with histopathology in order to group tumors into more biologically and molecularly defined entities. As such, tumor classification is significantly improved through better characterized natural histories. These changes have particularly important implications for gliomas. For the first time, adult- and pediatric-type gliomas are classified separately on the basis of differences in molecular pathogenesis and prognosis. Furthermore, the previous broad category of adult-type diffuse gliomas has been consolidated into 3 types: astrocytoma, isocitrate dehydrogenase (IDH) mutant; oligodendroglioma, IDH mutant and 1p/19q codeleted; and glioblastoma, IDH wild type. These major changes are driven by IDH mutation status and include the restriction of the diagnosis of glioblastoma to tumors that are IDH wild type; the reclassification of tumors previously diagnosed as IDH-mutated glioblastomas as astrocytomas IDH mutated, grade 4; and the requirement for the presence of IDH mutations to classify tumors as astrocytomas or oligodendrogliomas. CONCLUSIONS AND RELEVANCE: The 2021 World Health Organization central nervous system tumor classification is a major advance toward improving the diagnosis of brain tumors. It will provide clinicians with more accurate guidance on prognosis and optimal therapy for patients and ensure that more homogenous patient populations are enrolled in clinical trials, potentially facilitating the development of more effective therapies. DOI: 10.1001/jamaoncol.2022.2844 PMID: 36006639 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35876874
1. Neuroradiology. 2023 Jan;65(1):41-54. doi: 10.1007/s00234-022-03015-7. Epub 2022 Jul 25. Imaging-based stratification of adult gliomas prognosticates survival and correlates with the 2021 WHO classification. Kamble AN(1)(2), Agrawal NK(3)(4), Koundal S(5), Bhargava S(6), Kamble AN(7), Joyner DA(8), Kalelioglu T(8), Patel SH(8), Jain R(9)(10). Author information: (1)University Hospitals Coventry & Warwickshire, Coventry, UK. [email protected]. (2)Deep Learning Institute of Radiological Sciences (DeLoRIS), Mumbai, India. [email protected]. (3)Deep Learning Institute of Radiological Sciences (DeLoRIS), Mumbai, India. (4)Max Super-Specialty Hospital, Mohali, India. (5)Department of Radiology, Institute of Nuclear Medicine & Allied Sciences (INMAS), New Delhi, India. (6)Sir Ganga Ram Hospital, Delhi, India. (7)Saraswati College of Engineering, Mumbai, India. (8)Department of Radiology, University of Virginia Health System, Charlottesville, VA, USA. (9)Department of Radiology, New York University Grossman School of Medicine, New York, NY, USA. (10)Department of Neurosurgery, New York University Grossman School of Medicine, New York, NY, USA. BACKGROUND: Because of the lack of global accessibility, delay, and cost-effectiveness of genetic testing, there is a clinical need for an imaging-based stratification of gliomas that can prognosticate survival and correlate with the 2021-WHO classification. METHODS: In this retrospective study, adult primary glioma patients with pre-surgery/pre-treatment MRI brain images having T2, FLAIR, T1, T1 post-contrast, DWI sequences, and survival information were included in TCIA training-dataset (n = 275) and independent validation-dataset (n = 200). A flowchart for imaging-based stratification of adult gliomas(IBGS) was created in consensus by three authors to encompass all adult glioma types. Diagnostic features used were T2-FLAIR mismatch sign, central necrosis with peripheral enhancement, diffusion restriction, and continuous cortex sign. Roman numerals (I, II, and III) denote IBGS types. Two independent teams of three and two radiologists, blinded to genetic, histology, and survival information, manually read MRI into three types based on the flowchart. Overall survival-analysis was done using age-adjusted Cox-regression analysis, which provided both hazard-ratio (HR) and area-under-curve (AUC) for each stratification system(IBGS and 2021-WHO). The sensitivity and specificity of each IBSG type were analyzed with cross-table to identify the corresponding 2021-WHO genotype. RESULTS: Imaging-based stratification was statistically significant in predicting survival in both datasets with good inter-observer agreement (age-adjusted Cox-regression, AUC > 0.5, k > 0.6, p < 0.001). IBGS type-I, type-II, and type-III gliomas had good specificity in identifying IDHmut 1p19q-codel oligodendroglioma (training - 97%, validation - 85%); IDHmut 1p19q non-codel astrocytoma (training - 80%, validation - 85.9%); and IDHwt glioblastoma (training - 76.5%, validation- 87.3%) respectively (p-value < 0.01). CONCLUSIONS: Imaging-based stratification of adult diffuse gliomas predicted patient survival and correlated well with 2021-WHO glioma classification. © 2022. The Author(s), under exclusive licence to Springer-Verlag GmbH Germany, part of Springer Nature. DOI: 10.1007/s00234-022-03015-7 PMID: 35876874 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/36299937
1. Cureus. 2022 Sep 22;14(9):e29457. doi: 10.7759/cureus.29457. eCollection 2022 Sep. T2-Fluid-Attenuated Inversion Recovery (FLAIR) Mismatch as a Novel Specific MRI Marker for Adult Low-Grade Glioma (LGG): A Case Report. Slaghour RM(1), Almarshedi RA(2), Alzahrani AM(3), Albadr F(4). Author information: (1)Medicine, King Abdulaziz University Faculty of Medicine, Jeddah, SAU. (2)Medicine, University of Hail College of Medicine, Hail, SAU. (3)Medicine, King Saud Medical City, King Saud University, Riyadh, SAU. (4)Radiology and Medical Imaging/Neuroradiology, King Saud Medical City, King Saud University, Riyadh, SAU. Astrocytic tumors are primary central nervous system tumors. They are the most common tumors arising from glial cells. In the new WHO classification 2021, adult-type diffuse astrocytic gliomas subdivide into isocitrate dehydrogenase (IDH)-mutant astrocytoma, IDH-mutant and 1p/19q-codeleted oligodendroglioma, and IDH-wildtype glioblastoma. The T2-fluid-attenuated inversion recovery (FLAIR) mismatch sign describes the MRI appearance of IDH-mutant astrocytoma, it is considered a highly specific radiogenomic signature for diffuse astrocytoma, as opposed to other lower-grade. MRI is the first and most accurate diagnostic tool for low-grade gliomas (LGGs). It is particularly helpful in distinguishing a diffuse astrocytoma from an oligodendroglioma that will not demonstrate T2-FLAIR mismatch. The tumor displays a hyperintense signal on T2-weighted images and a hypointense signal on T2-weighted FLAIR images, which distinguishes it from other types of diffuse gliomas. We report a case of a 29-year-old female patient who was diagnosed with IDH-mutant 1p/19q-non-codeleted diffuse astrocytoma based on MRI T-2 FLAIR mismatch sign, which is confirmed by the molecular analysis in the pathology lab. Our aim of this report is to confirm the power of the MRI findings in the diagnosis of glioma genotypes and to assess neurosurgeons in the preoperative surgical planning. Copyright © 2022, Slaghour et al. DOI: 10.7759/cureus.29457 PMCID: PMC9587756 PMID: 36299937 Conflict of interest statement: The authors have declared that no competing interests exist.
http://www.ncbi.nlm.nih.gov/pubmed/21889777
1. Rev Neurol (Paris). 2011 Oct;167(10):683-90. doi: 10.1016/j.neurol.2011.07.006. Epub 2011 Sep 1. Histomolecular classification of adult diffuse gliomas: the diagnostic value of immunohistochemical markers. Figarella-Branger D(1), Maues de Paula A, Colin C, Bouvier C. Author information: (1)Service d'anatomie pathologique et de neuropathologie, hôpital de la Timone, Assistance publique-Hôpitaux de Marseille, rue Saint-Pierre, Marseille cedex 05, France. [email protected] Adult gliomas are most often infiltrative. The World Health Organization (WHO) has classed them into three major groups according to the presomptive cell of origin: astrocytoma, oligodendroglioma and mixed oligoastrocytoma. Depending on the presence or absence of a small number of signs of anaplasia (mitosis, nuclear atypia, cell density, microvascular proliferation and necrosis) the WHO distinguishes grade II (LGG), III (anaplastic), and IV (glioblastomas, GBM). Mutation in the isocitrate deshydrogenase I and II (IDH1 and 2) genes distinguishes grade II, III and secondary GBM from primary GBM. Moreover two additional genetic alterations are recorded in grade II and III gliomas: TP53 mutations that characterize astrocytomas and 1p19q codeletion (as the result of t(1;19)(q10;p10) translocation) recorded in oligodendrogliomas. Mixed gliomas, the most non-reproducible category, share with astrocytomas and oligodendrogliomas the same genetic alterations. Interestingly TP53 mutation (p53+) and 1p19q codeletion (1p19q+) are mutually exclusive and involve IDH mutated (IDH+) glial precursor cells. According to IDH, TP53, and 1p19q status, four major subtypes of LGG are recorded: IDH+/p53-/1p19q-, IDH+/p53+/1p19q-, IDH+/p53-/1p19q+ and triple negative, this last subgroup having the worst prognosis. Interestingly, p53 expression and internexin alpha (INA) expression can replace to some extent TP53 mutation and 1p19 codeletion, respectively. Moreover the antibody directed against the IDH1R132H isoform is highly specific. Because this mutation is the most frequent it is sufficient to assess IDH status in more than 80% of grade II and III gliomas. Taken together these three immunohistochemical markers are contribute greatly to the classification of gliomas and should be tested routinely as diagnostic markers. Finally, although GBM are genetically heterogeneous, the vast majority display EGFR amplification, often associated with EGFR expression, which can be helpful for diagnosis in certain cases. Copyright © 2011 Elsevier Masson SAS. All rights reserved. DOI: 10.1016/j.neurol.2011.07.006 PMID: 21889777 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34729052
1. J Carcinog. 2021 Oct 11;20:20. doi: 10.4103/jcar.jcar_17_21. eCollection 2021. Molecular classification and stratification of adult diffuse gliomas: A tertiary care center study. Anand N(1), Husain N(1), Varshney R(1), Malhotra KP(1), Kaif M(2). Author information: (1)Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India. (2)Department of Neurosurgery, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, Uttar Pradesh, India. BACKGROUND: Diffuse gliomas in the adult population are the most common primary central nervous system (CNS) tumors. The World Health Organization incorporated isocitrate dehydrogenase (IDH) mutations and 1p/19q co-deletion with histopathological features into an "integrated diagnosis" in the revised classification of tumors of CNS. These molecular subgroups of diffuse gliomas are found to stratify patients into prognostically distinct groups better than the histological classification. The objectives of the current study were to assess the frequency of IDH mutation, ATRX expression loss, p53 overexpression, and 1p/19q co-deletion detection in adult diffuse gliomas (Grade II, III, and IV) and to correlate them with clinicopathological and histopathological features. MATERIALS AND METHODS: The current study was a tertiary care hospital-based retrospective case series of 112 cases of adult diffuse gliomas. Immunohistochemistry (IHC)-based molecular detection was performed for IDH-1, ATRX, and p53 and fluorescent in situ hybridization (FISH) was performed for 1p/19q co-deletion detection. RESULTS: IDH-1 mutation was present in 30.4% (n = 34/112) cases, ATRX expression was lost in 18% (n = 19/104) cases, p53 was mutated in 39.3% (n = 42/107) cases and 1p19q was co-deleted in 25% (n = 4/16) cases. In the IDH1 mutant cases, with retained ATRX, FISH for 1p/19q co-deletion was performed and was co-deleted in four cases. CONCLUSION: The results of the present study indicate that IHC including IDH1/2, ATRX, and p53 is useful for the molecular classification of diffuse gliomas, which could be useful for the evaluation of prognosis, especially Grade III and II. Although the immunohistochemical approach does not replace genetic testing completely, it is a practical and powerful means of assessing molecular genetic changes. IDH mutations are the established markers of better prognosis in diffuse gliomas. Copyright: © 2021 Journal of Carcinogenesis. DOI: 10.4103/jcar.jcar_17_21 PMCID: PMC8531577 PMID: 34729052 Conflict of interest statement: There are no conflicts of interest.
http://www.ncbi.nlm.nih.gov/pubmed/32640746
1. Cancers (Basel). 2020 Jul 6;12(7):1817. doi: 10.3390/cancers12071817. Beyond IDH-Mutation: Emerging Molecular Diagnostic and Prognostic Features in Adult Diffuse Gliomas. Mirchia K(1), Richardson TE(1). Author information: (1)Department of Pathology, State University of New York, Upstate Medical University, Syracuse, NY 13210, USA. Diffuse gliomas are among the most common adult central nervous system tumors with an annual incidence of more than 16,000 cases in the United States. Until very recently, the diagnosis of these tumors was based solely on morphologic features, however, with the publication of the WHO Classification of Tumours of the Central Nervous System, revised 4th edition in 2016, certain molecular features are now included in the official diagnostic and grading system. One of the most significant of these changes has been the division of adult astrocytomas into IDH-wildtype and IDH-mutant categories in addition to histologic grade as part of the main-line diagnosis, although a great deal of heterogeneity in the clinical outcome still remains to be explained within these categories. Since then, numerous groups have been working to identify additional biomarkers and prognostic factors in diffuse gliomas to help further stratify these tumors in hopes of producing a more complete grading system, as well as understanding the underlying biology that results in differing outcomes. The field of neuro-oncology is currently in the midst of a "molecular revolution" in which increasing emphasis is being placed on genetic and epigenetic features driving current diagnostic, prognostic, and predictive considerations. In this review, we focus on recent advances in adult diffuse glioma biomarkers and prognostic factors and summarize the state of the field. DOI: 10.3390/cancers12071817 PMCID: PMC7408495 PMID: 32640746 Conflict of interest statement: The authors have no conflict of interest to declare.
http://www.ncbi.nlm.nih.gov/pubmed/25744348
1. Neurol Med Chir (Tokyo). 2015;55(1):28-37. doi: 10.2176/nmc.ra.2014-0229. Epub 2014 Dec 20. Pathology and genetics of diffuse gliomas in adults. Komori T(1). Author information: (1)Department of Laboratory Medicine and Pathology (Neuropathology), Tokyo Metropolitan Neurological Hospital. Republished in Neurol Med Chir (Tokyo). 2015;55 Suppl 1:28-37. The current World Health Organization (WHO) classification of tumors of the central nervous system (CNS) is essentially a lineage-oriented classification based on a presumable developmental tree of CNS. A four-tiered WHO grading scheme has been successfully applied to a spectrum of diffusely infiltrative astrocytomas, but it is not fully applicable to other gliomas, including oligodendrogliomas and ependymomas. Recent genetic studies have revealed that the major categories of gliomas, such as circumscribe astrocytomas, infiltrating astrocytomas/oligodendrogliomas, and glioblastoma, roughly correspond to major genetic alterations, including isocitrate dehydrogenases (IDHs) 1/2 mutations, TP53 mutations, co-deletion of chromosome arms 1p/19q, and BRAF mutation/fusion. These genetic alterations are clinically significant in terms of the response to treatment(s) and/or the prognosis. It is, thus, rational that future classification of gliomas should be based on genotypes, rather than phenotypes, although the genetic features of each tumor are not sufficiently understood at present to draw a complete map of the gliomas, and genetic testing is not yet available worldwide, particularly in Asian and African countries. This review summarizes the current concepts of the WHO classification, as well as the current understanding of the major genetic alterations in glioma and the potential use of these alterations as diagnostic criteria. DOI: 10.2176/nmc.ra.2014-0229 PMCID: PMC4533400 PMID: 25744348 [Indexed for MEDLINE] Conflict of interest statement: Conflicts of Interest Disclosure The author has no conflict of interest to declare.
http://www.ncbi.nlm.nih.gov/pubmed/25641008
1. Eur J Radiol. 2015 Apr;84(4):709-20. doi: 10.1016/j.ejrad.2014.12.025. Epub 2015 Jan 7. Imaging of adult brainstem gliomas. Purohit B(1), Kamli AA(2), Kollias SS(2). Author information: (1)Institute for Neuroradiology, University Hospital of Zurich, Frauenklinikstrasse 10, 8091 Zurich, Switzerland. Electronic address: [email protected]. (2)Institute for Neuroradiology, University Hospital of Zurich, Frauenklinikstrasse 10, 8091 Zurich, Switzerland. Brainstem gliomas (BSGs) are uncommon in adults accounting for about 2% of all intracranial neoplasms. They are often phenotypically low-grade as compared to their more common paediatric counterparts. Since brainstem biopsies are rarely performed, these tumours are commonly classified according to their MR imaging characteristics into 4 subgroups: (a) diffuse intrinsic low-grade gliomas, (b) enhancing malignant gliomas, (c) focal tectal gliomas and (d) exophytic gliomas/other subtypes. The prognosis and treatment is variable for the different types and is almost similar to adult supratentorial gliomas. Radiotherapy (RT) with adjuvant chemotherapy is the standard treatment of diffuse low-grade and malignant BSGs, whereas, surgical resection is limited to the exophytic subtypes. Review of previous literature shows that the detailed imaging of adult BSGs has not received significant attention. This review illustrates in detail the imaging features of adult BSGs using conventional and advanced MR techniques like diffusion weighted imaging (DWI), diffusion tensor imaging (DTI), MR perfusion weighted imaging (PWI), MR spectroscopy (MRS), as well as 18F-fluoro-ethyl-tyrosine positron emission tomography (18F-FET/PET). We have discussed the pertinent differences between childhood and adult BSGs, imaging mimics, prognostic factors and briefly reviewed the treatment options of these tumours. Copyright © 2015 Elsevier Ireland Ltd. All rights reserved. DOI: 10.1016/j.ejrad.2014.12.025 PMID: 25641008 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/28339700
1. Neuro Oncol. 2017 May 1;19(5):614-624. doi: 10.1093/neuonc/now277. A clinical perspective on the 2016 WHO brain tumor classification and routine molecular diagnostics. van den Bent MJ(1), Weller M(2), Wen PY(3), Kros JM(4), Aldape K(5), Chang S(6). Author information: (1)Department of Neurology and Brain Tumor Center, Erasmus MC Cancer Institute, Rotterdam, the Netherlands (2)Department of Neurology and Brain Tumor Center, University Hospital and University of Zurich, Zurich, Switzerland (3)Center for Neuro-Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts (4)Department of Pathology, Erasmus MC, Rotterdam, the Netherlands (5)Department of Pathology, Princess Margaret Hospital, Toronto, Canada (6)Division of Neuro-Oncology, Department of Neurological Surgery, University of California San Francisco,San Francisco, California Comment in Neuro Oncol. 2017 May 1;19(5):609-610. doi: 10.1093/neuonc/nox003. The 2007 World Health Organization (WHO) classification of brain tumors did not use molecular abnormalities as diagnostic criteria. Studies have shown that genotyping allows a better prognostic classification of diffuse glioma with improved treatment selection. This has resulted in a major revision of the WHO classification, which is now for adult diffuse glioma centered around isocitrate dehydrogenase (IDH) and 1p/19q diagnostics. This revised classification is reviewed with a focus on adult brain tumors, and includes a recommendation of genes of which routine testing is clinically useful. Apart from assessment of IDH mutational status including sequencing of R132H-immunohistochemistry negative cases and testing for 1p/19q, several other markers can be considered for routine testing, including assessment of copy number alterations of chromosome 7 and 10 and of TERT promoter, BRAF, and H3F3A mutations. For "glioblastoma, IDH mutated" the term "astrocytoma grade IV" could be considered. It should be considered to treat IDH wild-type grades II and III diffuse glioma with polysomy of chromosome 7 and loss of 10q as glioblastoma. New developments must be more quickly translated into further revised diagnostic categories. Quality control and rapid integration of molecular findings into the final diagnosis and the communication of the final diagnosis to clinicians require systematic attention. © The Author(s) 2017. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: [email protected]. DOI: 10.1093/neuonc/now277 PMCID: PMC5464438 PMID: 28339700 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/30148717
1. Curr Opin Oncol. 2018 Nov;30(6):375-382. doi: 10.1097/CCO.0000000000000482. Molecular classification of adult gliomas: recent advances and future perspectives. Barritault M(1)(2)(3), Meyronet D(4)(2)(3), Ducray F(5)(2)(3). Author information: (1)Service de Biologie Moléculaire. (2)Université Claude Bernard Lyon 1. (3)Department of Cancer Cell Plasticity, Cancer Research Centre of Lyon, INSERM U1052, CNRS UMR5286, Lyon, France. (4)Service de Neuropathologie. (5)Service de Neuro-Oncologie, Hospices Civils de Lyon, Groupe Hospitalier Est, Lyon, Cedex. PURPOSE OF REVIEW: This review summarizes recent advances in the molecular classification of adult gliomas. RECENT FINDINGS: According to the 2016 WHO classification, five main molecular subgroups of adult diffuse gliomas can be distinguished based on the 1p/19q codeletion, isocitrate dehydrogenase (IDH), and histone H3.3 mutation status. In the future, this classification may be further refined based on the integration of additional biomarkers, in particular CDKN2A/B homozygous deletion in IDH-mutant astrocytomas, TERT promoter mutations, EGFR amplification, chromosome 7 gain and chromosome 10 loss in IDH-wildtype astrocytomas, and FGFR1 mutations in midline gliomas. Histone H3.3 G34R/V defines a distinct subgroup of hemispheric IDH-wildtype high-grade gliomas occurring in young patients and FGFR gene fusions characterize a subgroup of IDH-wildtype glioblastomas that could benefit from specific treatment approaches. RNA sequencing may identify targetable gene fusions in circumscribed gliomas lacking classical BRAF alterations. In chordoid gliomas, recurrent PRKCA mutations could serve as a new diagnostic marker. Among comprehensive molecular analysis methods, DNA methylation profiling appears as a particularly powerful approach to identify new molecular subgroups of gliomas and to classify difficult cases. SUMMARY: The classification of adult gliomas may be improved by the integration of additional biomarkers and/or by comprehensive molecular analysis, in particular DNA methylation profiling. The most relevant approach, however, remains to be established. DOI: 10.1097/CCO.0000000000000482 PMID: 30148717 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34788571
1. Nucleic Acid Ther. 2022 Feb;32(1):29-39. doi: 10.1089/nat.2021.0043. Epub 2021 Nov 17. Long-Term Safety and Efficacy Data of Golodirsen in Ambulatory Patients with Duchenne Muscular Dystrophy Amenable to Exon 53 Skipping: A First-in-human, Multicenter, Two-Part, Open-Label, Phase 1/2 Trial. Servais L(1)(2)(3), Mercuri E(4)(5), Straub V(6), Guglieri M(6), Seferian AM(1), Scoto M(7)(8), Leone D(5), Koenig E(9), Khan N(9), Dugar A(9), Wang X(9), Han B(9), Wang D(9), Muntoni F(7)(8); SKIP-NMD Study Group. Collaborators: Brusa C, Antonaci L, Brogna C, Merli L, Monforte M, Norcia G, Pane M, Ferrantini G, Dickson G, Morgan J, Sardone V, Akana C, Charleston JS, Desjardins CA, El-Husayni SH, Frank DE, Schnell FJ. Author information: (1)I-Motion Institute, Hôpital Armand Trousseau, Paris, France. (2)Division of Child Neurology, Centre de Références des Maladies Neuromusculaires, Department of Pediatrics, University Hospital Liège & University of Liège, Liège, Belgium. (3)MDUK Oxford Neuromuscular Centre, University of Oxford, Oxford, United Kingdom. (4)Pediatric Neurology Unit, Università Cattolica del Sacro Cuore Roma, Rome, Italy. (5)Nemo Clinical Centre, Fondazione Policlinico Universitario A Gemelli IRCCS, Rome, Italy. (6)John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom. (7)Dubowitz Neuromuscular Centre, University College London, Great Ormond Street Institute of Child Health, London, United Kingdom. (8)National Institute for Health Research Great Ormond Street Hospital Biomedical Research Centre, London, United Kingdom. (9)Sarepta Therapeutics, Inc., Cambridge, Massachusetts, USA. The aim of this Phase 1/2, 2-part, multicenter trial was to report clinical safety and efficacy of long-term golodirsen treatment among ambulatory patients with exon 53 skip-amenable Duchenne muscular dystrophy (DMD). Part 1 was a 12-week, randomized, double-blind, placebo-controlled, dose-titration study followed by 9-week safety review. Part 2 was a 168-week, open-label evaluation of golodirsen 30 mg/kg. Part 1 primary endpoint was safety. Part 2 primary endpoints were dystrophin protein expression and 6-minute walk test (6MWT); secondary endpoints were percent predicted forced vital capacity (FVC%p) and safety. Post hoc ambulation analyses used mutation-matched external natural history controls. All patients from Part 1 (golodirsen, n = 8; placebo, n = 4) plus 13 additional patients entered Part 2; 23 completed the study. Adverse events were generally mild, nonserious, and unrelated to golodirsen, with no safety-related discontinuations or deaths. Golodirsen increased dystrophin protein (16.0-fold; P < 0.001) and exon skipping (28.9-fold; P < 0.001). At 3 years, 6MWT change from baseline was -99.0 m for golodirsen-treated patients versus -181.4 m for external controls (P = 0.067), and loss of ambulation occurred in 9% versus 26% (P = 0.21). FVC%p declined 8.4% over 3 years in golodirsen-treated patients, comparing favorably with literature-reported rates. This study provides evidence for golodirsen biologic activity and long-term safety in a declining DMD population and suggests functional benefit versus external controls. Clinical Trial Registration number: NCT02310906. DOI: 10.1089/nat.2021.0043 PMCID: PMC8817703 PMID: 34788571 [Indexed for MEDLINE] Conflict of interest statement: L.S. has served on advisory boards for Sarepta Therapeutics, Inc.; E.M. has received consultant fees from Sarepta Therapeutics, Inc.; V.S. has received speaker honoraria from Sanofi Genzyme, is or has recently been on advisory boards for Audentes Therapeutics, Biogen, Exonics Therapeutics/Vertex, Novartis, Roche, Sarepta Therapeutics, Inc., and Wave Therapeutics, and has research collaborations with Sanofi Genzyme and Ultragenyx; M.G. has received speaker honoraria from Sarepta Therapeutics, Inc., is on advisory boards for Pfizer, has research collaboration with Sarepta Therapeutics, Inc., and is the Chair of the VBP15-004 study but does not have any financial interest with ReveraGen; A.M.S., M.S., and D.L. have nothing to disclose; E.K., N.K., A.D., X.W., B.H., and D.W. are or have been employees of Sarepta Therapeutics, Inc.; F.M. has received consultant fees and speaker honoraria from Sarepta Therapeutics, Inc., and is supported by the NIHR Great Ormond Street Hospital Biomedical Research Centre.
http://www.ncbi.nlm.nih.gov/pubmed/33833444
1. Nat Rev Drug Discov. 2021 Jun;20(6):476-488. doi: 10.1038/s41573-021-00175-8. Epub 2021 Apr 8. Industrializing engineered autologous T cells as medicines for solid tumours. Britten CM(1)(2), Shalabi A(3), Hoos A(4). Author information: (1)Oncology R&D, GlaxoSmithKline, Stevenage, UK. (2)Immatics Biotechnologies, Munich, Germany. (3)Oncology R&D, GlaxoSmithKline, Philadelphia, PA, USA. (4)Oncology R&D, GlaxoSmithKline, Philadelphia, PA, USA. [email protected]. Cell therapy is one of the fastest growing areas in the pharmaceutical industry, with considerable therapeutic potential. However, substantial challenges regarding the utility of these therapies will need to be addressed before they can become mainstream medicines with applicability similar to that of small molecules or monoclonal antibodies. Engineered T cells have achieved success in the treatment of blood cancers, with four chimeric antigen receptor (CAR)-T cell therapies now approved for the treatment of B cell malignancies based on their unprecedented efficacy in clinical trials. However, similar results have not yet been achieved in the treatment of the much larger patient population with solid tumours. For cell therapies to become mainstream medicines, they may need to offer transformational clinical effects for patients and be applicable in disease settings that remain unaddressed by simpler approaches. This Perspective provides an industry perspective on the progress achieved by engineered T cell therapies to date and the opportunities and current barriers for accessing broader patient populations, and discusses the solutions and new development strategies required to fully industrialize the therapeutic potential of engineered T cells as medicines. DOI: 10.1038/s41573-021-00175-8 PMID: 33833444 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/36401026
1. Methods Mol Biol. 2023;2587:107-124. doi: 10.1007/978-1-0716-2772-3_6. Restoring Dystrophin Expression by Skipping Exons 6 and 8 in Neonatal Dystrophic Dogs. Shah MNA(1), Yokota T(2)(3). Author information: (1)Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada. (2)Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada. [email protected]. (3)Muscular Dystrophy Canada Research Chair, Edmonton, AB, Canada. [email protected]. Duchenne muscular dystrophy (DMD) is caused by the mutations in the DMD gene resulting in no dystrophin production. Skipping DMD exons using phosphorodiamidate morpholino oligomers (PMOs) is an emerging treatment strategy that can restore the reading frame of the mutated gene and produce truncated but functional dystrophin protein. To date, four PMOs, including eteplirsen, casimersen, viltolarsen, and golodirsen, have been conditionally approved by the FDA for the treatment of DMD. Since degeneration of muscle fibers and irreversible fibrosis occur from childhood, the earlier treatment is preferred. The canine X-linked muscular dystrophy in Japan (CXMDj), a dog model of DMD, produces no dystrophin and exhibits a severe phenotype similar to human patients from early childhood. As such, CXMDj, which harbors a splice site mutation in intron 6, is a useful model for examining the long-term effects of early PMO treatment. In this chapter, we describe the systemic delivery of a cocktail of four PMOs that can successfully induce multiple exon skipping (exons 6-9) in neonatal dystrophic dogs. We also describe the procedures to evaluate the efficacy and toxicity, including clinical grading of dystrophic dogs, ELISA-based quantification of PMOs, histology, RT-PCR, and western blotting. © 2023. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature. DOI: 10.1007/978-1-0716-2772-3_6 PMID: 36401026 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32026421
1. Drugs. 2020 Feb;80(3):329-333. doi: 10.1007/s40265-020-01267-2. Golodirsen: First Approval. Heo YA(1). Author information: (1)Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand. [email protected]. Golodirsen (Vyondys 53™), an antisense oligonucleotide of the phophorodiamidate morpholino oligomer (PMO) subclass designed to induce exon 53 skipping, has been developed by Sarepta Therapeutics for the treatment of Duchenne muscular dystrophy (DMD). In December 2019, intravenous golodirsen received its first global approval in the USA for the treatment of DMD in patients with a confirmed mutation of the DMD gene that is amenable to exon 53 skipping, based on positive results from a phase I/II clinical trial. Golodirsen is in phase III clinical development for the treatment of DMD worldwide. This article summarizes the milestones in the development of golodirsen leading to this first approval for DMD. DOI: 10.1007/s40265-020-01267-2 PMID: 32026421 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/33025945
1. Drugs Today (Barc). 2020 Aug;56(8):491-504. doi: 10.1358/dot.2020.56.8.3159186. Golodirsen for Duchenne muscular dystrophy. Anwar S(1), Yokota T(2). Author information: (1)Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada. (2)Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada; The Friends of Garrett Cumming Research and Muscular Dystrophy Canada, HM Toupin Neurological Science Research Chair, Edmonton, Alberta, Canada. [email protected]. Duchenne muscular dystrophy (DMD) is a life-shortening X-linked genetic disorder characterized by progressive wasting and weakening of muscles in boys. Loss-of-function mutations in the DMD gene, which codes for dystrophin, lead to this disease. The majority of mutations in this gene result in the exclusion of one or more exons from the transcript, eventually causing the remaining exons not to fit together correctly (i.e., out-of-frame mutations). Antisense oligonucleotides, e.g., phosphorodiamidate morpholino oligomers (PMOs), can induce therapeutic exon skipping during pre-mRNA processing to restore the reading frame of the primary transcript of DMD. As a result, truncated but partially functional dystrophin is produced, potentially slowing down the disease progression. Golodirsen is a provisionally approved PMO-based drug for approx. 8% of all DMD patients amenable to exon 53 skipping. This article summarizes golodirsen's pharmacology, efficacy and safety information. It also discusses some controversies that golodirsen met after the approval. Copyright 2020 Clarivate Analytics. DOI: 10.1358/dot.2020.56.8.3159186 PMID: 33025945 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/16700618
1. Technol Cancer Res Treat. 2006 Jun;5(3):215-29. doi: 10.1177/153303460600500305. Molecular diagnostics as a tool to personalize treatment in adult glioma patients. Jeuken JW(1), van der Maazen RW, Wesseling P. Author information: (1)Department of Pathology, Nijmegen Centre for Molecular Life Sciences (NCMLS), Radboud University Nijmegen, Medical Centre, Nijmegen, The Netherlands. [email protected] Gliomas, the most frequent primary brain tumors in humans, form a heterogeneous group, encompassing many different histological types and malignancy grades. Within this group, the diffuse infiltrative gliomas are by far the most common in adults. The major representatives in this subgroup are the diffuse astrocytic, oligodendroglial, and mixed oligo-astrocytic tumors. Especially in these diffuse gliomas, the role of molecular diagnostics is rapidly increasing. After summarizing the most relevant genetic aberrations and pathways in these tumors detected up till now, this review will discuss the clinical relevance of this information. Several molecular markers have been identified in diffuse gliomas that carry diagnostic and prognostic information. In addition, some of these and other markers predict the response of these gliomas to particular (chemo)therapeutic approaches. The techniques used to obtain this molecular information, as well as the advantages and disadvantages of the different techniques will be discussed. Finally, future perspectives will be presented with regard to the contribution of molecular diagnostics to tailor-made therapy in glioma patients. DOI: 10.1177/153303460600500305 PMID: 16700618 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35330989
1. J Inflamm Res. 2022 Mar 18;15:1935-1941. doi: 10.2147/JIR.S356316. eCollection 2022. Application of Baricitinib in Dermatology. Zhang J(1), Qi F(1), Dong J(1), Tan Y(1), Gao L(2), Liu F(1). Author information: (1)Department of Dermatology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, People's Republic of China. (2)China CDC Key Laboratory of Radiological Protection and Nuclear Emergency, National Institute for Radiological Protection, Chinese Centre for Disease Control and Prevention, Beijing, 100088, People's Republic of China. There are four JAK subtypes: JAK1, JAK2, JAK3, and tyrosine kinase 2 (TYK2). Small molecule Janus tyrosine kinase (JAK) inhibitors can inhibit a variety of pro-inflammatory cytokines. Baricitinib is the first generation of JAK1/2 inhibitor targeting the ATPase of JAK, which blocks the intracellular transmission of cytokines through JAK-STATs. Thus far, it has been approved for the treatment of rheumatoid arthritis (RA); however, an increasing number of studies have suggested that baricitinib can be used to treat dermatological diseases, such as atopic dermatitis (AD), psoriasis, vitiligo, and alopecia areata. Baricitinib can be a new choice for the treatment of dermatological diseases, which cannot be treated with conventional drugs. We reviewed the application, efficacy, side effects, precautions, limitations and prospect of baricitinib in atopic dermatitis, psoriasis, vitiligo and alopecia areata (AA) in recent 5 years including clinical trials and case reports. Among them, the application in the field of alopecia areata is the most encouraging, and we reviewed the mechanism in detail. © 2022 Zhang et al. DOI: 10.2147/JIR.S356316 PMCID: PMC8939862 PMID: 35330989 Conflict of interest statement: The authors report no conflicts of interest in this work.
http://www.ncbi.nlm.nih.gov/pubmed/35334197
1. N Engl J Med. 2022 May 5;386(18):1687-1699. doi: 10.1056/NEJMoa2110343. Epub 2022 Mar 26. Two Phase 3 Trials of Baricitinib for Alopecia Areata. King B(1), Ohyama M(1), Kwon O(1), Zlotogorski A(1), Ko J(1), Mesinkovska NA(1), Hordinsky M(1), Dutronc Y(1), Wu WS(1), McCollam J(1), Chiasserini C(1), Yu G(1), Stanley S(1), Holzwarth K(1), DeLozier AM(1), Sinclair R(1); BRAVE-AA Investigators. Collaborators: Harada K, Ishii N, Ito T, Tateishi C, Choi GS, Huh CH, Jang YH, Kang H, Kim B, Kim MB, Kwon O, Lee YW, Lee Y, Lew BL, Yun SK, Barragan Estudillo Z, Herz M, Lopez Lozano H, Poletti Vazquez E, Rodriguez Acosta ED, Rodriguez Castellanos M, Villanueva Quintero DG, Bergfeld W, Blauvelt A, Browning J, Callender V, Cather J, Cohen D, Cohen S, Colavincenzo M, Davis S, Deck K, Ferris L, Forman S, Gonzalez-Chavez J, Gottlieb S, Greenstein D, Greiling T, Hordinsky M, Jarell A, Jenkin P, Johnson S, Kaffenberger J, King B, Ko J, Korman N, Krueger G, Kwong P, Lain E, Wine Lee L, Lee P, Ling M, Madkan V, Maverakis E, Mayo T, McFalda W, Mercurio M, Mesinkovska N, Miller J, Mostaghimi A, Nahm W, Owen C, Roberts J, Schmieder G, Senna M, Smith S, Sofen H, Taylor S, Teller C, Tosti A, Trovato A, Tyring S, Vendrell-Benito P, Wolfe J, Cappetta E, Castro V, Cecilia MN, Galimberti ML, Luna P, Marini M, Parra V, Abdulla EAK, Foley P, Gebauer K, Rubel D, Sidhu S, Sinclair R, Spelman L, Azulay-Abulafia L, Blessmann Weber M, Chicralla da Rocha N, Gonzaga da Cunha M, Kakizaki P, Lazzarini R, Magalhaes R, Romanelli Tiburcio Alves C, Yang Q, Zhang CL, Goldsmith T, Lyakhovitsky A, Martinez H, Ramon M, Valdman-Grinshpoun Y, Ziv M, Zlotogorski A, Kinoshita-Ise M, Komiyama E, Sato K, Shimomura Y, Ueki R, Choi JW, Kim DY, Lee SH, Park BC, Park H, Huang YH, Lai PJ, Tsai TF, Yang CC, Boyce B, Poynot Couvillion M, Dabiri G, Dhawan S, Filipof N, Fretzin S, Hamzavi F, Hake Harris H, Hussain I, Kelly J, Kempers S, Kerdel F, Krell JM, Laquer V, LaTowsky B, Lee M, Lockshin B, Lugo-Somolinos A, Martinez M, Mirmirani P, Moss J, Nasir A, Nixon D, Nunez L, Osman L, Pariser D, Rich P, Rosmarin D, Schwartz J, Serrao R, Ston M, Truett A, Zirwas M. Author information: (1)From the Yale School of Medicine, New Haven, CT (B.K.); the Department of Dermatology, Kyorin University Faculty of Medicine, Tokyo (M.O.); the Seoul National University College of Medicine, Seoul, South Korea (O.K.); the Department of Dermatology, Hadassah Medical Center, Hebrew University of Jerusalem, Faculty of Medicine, Jerusalem, Israel (A.Z.); Stanford University School of Medicine, Stanford (J.K.), and the University of California Irvine, Irvine (N.A.M.) - both in California; the Department of Dermatology, University of Minnesota Medical School, Minneapolis (M.H.); Eli Lilly, Indianapolis (Y.D., W.-S.W., J.M., C.C., G.Y., S.S., K.H., A.M.D.); and Sinclair Dermatology, Melbourne, VIC, Australia (R.S.). Comment in N Engl J Med. 2022 May 5;386(18):1751-1752. doi: 10.1056/NEJMe2203440. Dermatol Ther. 2022 Dec;35(12):e15845. doi: 10.1111/dth.15845. Br J Dermatol. 2023 Feb 10;188(2):163. doi: 10.1093/bjd/ljac080. BACKGROUND: Alopecia areata is an autoimmune condition characterized by rapid hair loss in the scalp, eyebrows, and eyelashes, for which treatments are limited. Baricitinib, an oral, selective, reversible inhibitor of Janus kinases 1 and 2, may interrupt cytokine signaling implicated in the pathogenesis of alopecia areata. METHODS: We conducted two randomized, placebo-controlled, phase 3 trials (BRAVE-AA1 and BRAVE-AA2) involving adults with severe alopecia areata with a Severity of Alopecia Tool (SALT) score of 50 or higher (range, 0 [no scalp hair loss] to 100 [complete scalp hair loss]). Patients were randomly assigned in a 3:2:2 ratio to receive once-daily baricitinib at a dose of 4 mg, baricitinib at a dose of 2 mg, or placebo. The primary outcome was a SALT score of 20 or less at week 36. RESULTS: We enrolled 654 patients in the BRAVE-AA1 trial and 546 in the BRAVE-AA2 trial. The estimated percentage of patients with a SALT score of 20 or less at week 36 was 38.8% with 4-mg baricitinib, 22.8% with 2-mg baricitinib, and 6.2% with placebo in BRAVE-AA1 and 35.9%, 19.4%, and 3.3%, respectively, in BRAVE-AA2. In BRAVE-AA1, the difference between 4-mg baricitinib and placebo was 32.6 percentage points (95% confidence interval [CI], 25.6 to 39.5), and the difference between 2-mg baricitinib and placebo was 16.6 percentage points (95% CI, 9.5 to 23.8) (P<0.001 for each dose vs. placebo). In BRAVE-AA2, the corresponding values were 32.6 percentage points (95% CI, 25.6 to 39.6) and 16.1 percentage points (95% CI, 9.1 to 23.2) (P<0.001 for each dose vs. placebo). Secondary outcomes for baricitinib at a dose of 4 mg but not at a dose of 2 mg generally favored baricitinib over placebo. Acne, elevated levels of creatine kinase, and increased levels of low- and high-density lipoprotein cholesterol were more common with baricitinib than with placebo. CONCLUSIONS: In two phase 3 trials involving patients with severe alopecia areata, oral baricitinib was superior to placebo with respect to hair regrowth at 36 weeks. Longer trials are required to assess the efficacy and safety of baricitinib for alopecia areata. (Funded by Eli Lilly under license from Incyte; BRAVE-AA1 and BRAVE-AA2 ClinicalTrials.gov numbers, NCT03570749 and NCT03899259.). Copyright © 2022 Massachusetts Medical Society. DOI: 10.1056/NEJMoa2110343 PMID: 35334197 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35774427
1. Chronic Dis Transl Med. 2022 Apr 18;8(2):134-144. doi: 10.1002/cdt3.23. eCollection 2022 Jun. Efficacy and safety of darbepoetin alfa injection replacing epoetin alfa injection for the treatment of renal anemia in Chinese hemodialysis patients: A randomized, open-label, parallel-group, noninferiority phase III trial. Liu B(1), Chen N(2), Zhao J(3), Yin A(4), Wu X(5), Xing C(6), Jiang G(7), Fu J(8), Wang M(9), Wang R(10), Niu J(11), Fu P(12), Ni Z(13), Hou F(14), Zhao J(15), Chen J(16), Chen Y(17), Shi W(18), Chen J(19), Li W(20), Xu G(21), Zhong L(22), Liu W(23), Ding G(24), Kondo Y(25), Yue C(26), Mei C(27). Author information: (1)Department of Nephrology Zhongda Hospital Southeast University Nanjing Jiangsu 210009 China. (2)Department of Nephrology Ruijin Hospital Shanghai Jiaotong University School of Medicine Shanghai 200025 China. (3)Department of Nephrology Xinqiao Hospital of Army Medical University Chongqing 400037 China. (4)Department of Nephrology First Affiliated Hospital of Xi'an Jiaotong University Xi'an Shaanxi 710061 China. (5)Department of Nephrology Southwest Hospital of Army Medical University Chongqing 400039 China. (6)Department of Nephrology Jiangsu Provincial Hospital Nanjing Jiangsu 210036 China. (7)Department of Nephrology Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine Shanghai 200092 China. (8)Department of Nephrology Guangzhou First People's Hospital Guangzhou Guangdong 510180 China. (9)Department of Nephrology Peking University People's Hospital Beijing 100044 China. (10)Department of Nephrology Shandong Provincial Hospital Jinan Shandong 250021 China. (11)Department of Nephrology, Shanghai Fifth People's Hospital Fudan University Shanghai 200240 China. (12)Department of Nephrology, West China Hospital Sichuan University Chengdu Sichuan 610041 China. (13)Department of Nephrology Renji Hospital Shanghai Jiaotong University School of Medicine Shanghai 200001 China. (14)Department of Nephrology Nanfang Hospital Southern Medical University Guangzhou Guangdong 510510 China. (15)Department of Nephrology The Second Hospital of Dalian Medical University Dalian Liaoning 116023 China. (16)Department of Nephrology, Huashan Hospital Fudan University Shanghai 200040 China. (17)Department of Nephrology Peking University First Hospital Beijing 100034 China. (18)Department of Nephrology Guangdong Provincial People's Hospital Guangzhou Guangdong 510080 China. (19)Department of Nephrology, The First Affiliated Hospital Zhejiang University School of Medicine Hangzhou Zhejiang 310003 China. (20)Department of Nephrology China-Japan Friendship Hospital Beijing 100029 China. (21)Department of Nephrology Tongji Hospital Tongji Medical College Huazhong University of Science and Technology Wuhan Hubei 430030 China. (22)Department of Nephrology The Second Affiliated Hospital of Chongqing Medical University Chongqing 400010 China. (23)Department of Nephrology, Beijing Friendship Hospital Capital Medical University Beijing 100050 China. (24)Department of Nephrology Renmin Hospital of Wuhan University Wuhan Hubei 430060 China. (25)R&D Planning Department, R&D Division Kyowa Kirin Co., Ltd. Tokyo 520-5292 Japan. (26)D&R Office, Kyowa Kirin China Pharmaceutical Co., Ltd. Shanghai 201203 China. (27)Department of Nephrology Shanghai Changzheng Hospital Shanghai 200003 China. BACKGROUND: This study was to explore the clinical efficacy and safety of darbepoetin alfa injection replacing epoetin alfa injection (recombinant human erythropoietin injection, rHuEPO) for the treatment of anemia associated with chronic kidney failure in Chinese patients undergoing hemodialysis. METHOD: This study was a multicenter, randomized, open-label, intergroup parallel control phase III noninferiority trial from April 19, 2013 to September 9, 2014 at 25 sites. In this study, the members of the darbepoetin alfa group underwent intravenous administration once per week or once every two weeks. The members of the control drug epoetin alfa group underwent intravenous administration two or three times per week. All subjects underwent epoetin alfa administration during the 8-week baseline period. After that, subjects were randomly assigned to the darbepoetin alfa group or epoetin alfa group. The noninferiority in the changes of the average Hb concentrations from the baseline to the end of the evaluation period (noninferiority threshold: -1.0 g/dl) was tested between the two treatments. The time-dependent hemoglobin (Hb) concentration and the maintenance rate of the target Hb concentration (the proportion of subjects with Hb concentrations between 10.0 and 12.0 g/dl) were also evaluated. Iron metabolism, including changes in the serum iron, total iron-binding capacity, ferritin, transferrin saturation, and comparisons of the dose adjustments between the two groups during the treatment period were analyzed further. Adverse events (AEs) were also observed and compared, and the safety was analyzed between the two treatment groups. The conversion rate switching from epoetin alfa to darbepoetin alfa was also discussed. SAS® software version 9.2 was used to perform all statistical analyses. Descriptive statistics were used for all efficacy, safety, and demographic variable analyses, including for the primary efficacy indicators. RESULTS: Four hundred and sixty-six patients were enrolled in this study, and ultimately 384 cases were analyzed for safety, including 267 cases in the darbepoetin alfa group and 117 cases in the epoetin alfa group. There were 211 cases in the per-protocol set, including 152 cases in the darbepoetin alfa group and 59 cases in the epoetin alfa group. The changes in the average Hb concentrations from the baseline to the end of the evaluation period were -0.07 and -0.15 g/dl in the darbepoetin alfa group and epoetin alfa group respectively. The difference between the two groups was 0.08 g/dl (95% confidence interval [CI]: -0.22 to 0.39), and the lower limit of the 95% CI was -0.22 > -1.0 g/dl. The average Hb concentrations of the two groups were 10.88-11.43 g/dl (darbepoetin alfa) and 10.91-11.38 g/dl (epoetin alfa) during the study period of Weeks 0-28, with the maintenance rates of the target Hb concentration ranging within 71%-87% and 78%-95% in the darbepoetin alfa group and epoetin alfa group respectively. During the period of comparison between the two groups, the incidence of AEs in the darbepoetin alfa group was 61.42%, while in the epoetin alfa group it was 56.41%. All of the adverse events and reactions in the study were those commonly associated with hemodialysis. CONCLUSION: The overall efficacy and safety of darbepoetin alfa for the treatment of Chinese renal anemia patients undergoing hemodialysis are consistent with those of epoetin alfa. © 2022 The Authors. Chronic Diseases and Translational Medicine published by John Wiley & Sons Ltd on behalf of Chinese Medical Association. DOI: 10.1002/cdt3.23 PMCID: PMC9215713 PMID: 35774427 Conflict of interest statement: Kyowa Kirin Co., Ltd. (KKC) and Kyowa Kirin China Pharmaceutical Co., Ltd (KKCN) participated in and approved the design and conduct of the study. Professor Wenge Li is a member of Chronic Diseases and Translational Medicine editorial board and is not involved in the peer review process of this article.
http://www.ncbi.nlm.nih.gov/pubmed/21877597
1. Vnitr Lek. 2011 Jul-Aug;57(7-8):631-4. [Anemia and chronic kidney failure]. [Article in Czech] Stepánková S(1). Author information: (1)Centrum kardiovaskulární a transplantacní chirurgie Brno. [email protected] Anaemia is a common manifestation ofa chronic kidney failure. It is caused by a relative shortage oferythropoetine (EPO) and iron deficite with its metabolism defect. The most important factor in the pathogenesis of iron metabolism defects is hepcidin. Hepcidin maintains the iron homeostasis in the organism. The therapy of renal anaemia is based on the iron substitution and erythropoiesis stimulating agents (ESA) application. The most common reasons for the resistance to ESA are (after iron deficiency) inflammation and malnutrition. PMID: 21877597 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/15467179
1. Perspect Biol Med. 2004 Autumn;47(4):575-89. doi: 10.1353/pbm.2004.0072. The role of anemia in congestive heart failure and chronic kidney insufficiency: the cardio renal anemia syndrome. Silverberg DS(1), Wexler D, Iaina A. Author information: (1)Department of Nephrology, Tel Aviv Medical Center, Weizman 6, Israel. [email protected] Anemia is a major problem in patients with chronic kidney insufficiency. The development of recombinant human erythropoietin has enabled physicians to correct this anemia. Although anemia has not been considered to be a common or important contributor to congestive heart failure, anemia of any cause can lead to cardiac damage and eventually congestive heart failure. Our joint renal-cardiac heart failure team found that anemia was indeed very common in congestive heart failure and was associated with severe, medication-resistant cardiac failure. Correction of the anemia with erythropoietin and intravenous iron led to a marked improvement in patients' functional status and their cardiac function, and to a marked fall in the need for hospitalization and for high-dose diuretics; renal function usually improved or at least stabilized. Subsequent investigations by others have confirmed many of our observations. We call this interrelationship between congestive heart failure, chronic kidney insufficiency, and anemia the Cardio-Renal Anemia syndrome. Treatment of the anemia in congestive heart failure may prove vital in preventing progression of both the heart failure and the associated renal disease. DOI: 10.1353/pbm.2004.0072 PMID: 15467179 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/16623112
1. Przegl Lek. 2005;62 Suppl 2:19-21. [Anemia in kidney diseases as a cardiovascular risk factor]. [Article in Polish] Myśliwiec M(1), Zalewski G. Author information: (1)Klinika Nefrologii i Transplantologii z Ośrodkiem Dializ Akademii Medycznej w Białymstoku. [email protected] Cardiovascular mortality is increased in chronic kidney diseases. Patients' survival correlates with hemoglobin concentration. Anemia significantly affects cardiovascular system, especially the heart. Heart failure, anemia and chronic renal failure are mutually related. Recently, cardio-renal-anemia syndrome (CRAS) was defined. Iron replacement and EPO treatment can break the pathogenetic circle of events leading to heart failure. Increased hemoglobin concentration can also induce a positive impact on renal function. Early diagnosis and correction of anemia has a substantial role in prophylaxis of left ventricular hypertrophy in patients with chronic renal insufficiency. PMID: 16623112 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/16442920
1. Am Heart J. 2006 Feb;151(2):492-500. doi: 10.1016/j.ahj.2005.03.055. Kidney function and anemia as risk factors for coronary heart disease and mortality: the Atherosclerosis Risk in Communities (ARIC) Study. Astor BC(1), Coresh J, Heiss G, Pettitt D, Sarnak MJ. Author information: (1)Department of Epidemiology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA. [email protected] Comment in Am Heart J. 2006 Aug;152(2):e21. doi: 10.1016/j.ahj.2006.04.031. BACKGROUND: Kidney failure causes anemia and is associated with a very high risk of coronary heart disease (CHD). Mildly to moderately decreased kidney function is far more common and also is associated with an elevated prevalence of anemia and CHD risk. Recent data suggest an even higher risk of CHD when both conditions are present. METHODS: We investigated the association of kidney dysfunction and anemia with CHD events (fatal or nonfatal CHD or coronary revascularization procedures) and CHD and all-cause mortality over 12 years of follow-up in 14971 adults aged 45 to 64 years in the ARIC Study. Glomerular filtration rate (GFR) was estimated from calibrated serum creatinine using the MDRD Study equation (< 30 mL/min per 1.73 m2 excluded, n = 32). Anemia was defined as hemoglobin level < 13.5 g/dL in men (648/6746, 9.6%) and < 12 g/dL in women (1049/8225, 12.8%). RESULTS: The prevalence of anemia was progressively higher at lower estimated GFR < 75 mL/min per 1.73 m2 (both P < .001) for both men and women. A total of 1635 (10.9%) participants had a CHD event, 360 (2.4%) died of CHD, and 1722 (11.5%) died of any cause during follow-up. After adjustment for known risk factors, including diabetes, lipid levels, blood pressure, and use of antihypertensive medication, decreased kidney function was associated with a higher risk of recurrent CHD events and mortality from CHD and all causes. These associations were significantly stronger among participants with anemia. The adjusted relative hazards of all-cause mortality associated with moderately decreased versus normal kidney function (GFR 30-59 vs > or = 90 mL/min per 1.73 m2) were 1.7 (95% CI 1.3-2.2) in the absence of anemia and 3.5 (95% CI 2.4-5.1) in the presence of anemia (P interaction = .001). CONCLUSIONS: The combination of moderately decreased kidney function and anemia is associated with an increased risk of CHD events and mortality, emphasizing the need to identify individuals with these conditions and evaluate interventions to treat anemia and slow the progression of chronic kidney disease. DOI: 10.1016/j.ahj.2005.03.055 PMID: 16442920 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/17108343
1. N Engl J Med. 2006 Nov 16;355(20):2085-98. doi: 10.1056/NEJMoa065485. Correction of anemia with epoetin alfa in chronic kidney disease. Singh AK(1), Szczech L, Tang KL, Barnhart H, Sapp S, Wolfson M, Reddan D; CHOIR Investigators. Author information: (1)Renal Division, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA. [email protected] Comment in N Engl J Med. 2006 Nov 16;355(20):2144-6. doi: 10.1056/NEJMe068233. Nephrol Dial Transplant. 2007 Feb;22(2):309-12. doi: 10.1093/ndt/gfl824. N Engl J Med. 2007 Mar 1;356(9):956; author reply 958-9. doi: 10.1056/NEJMc063535. N Engl J Med. 2007 Mar 1;356(9):956-7; author reply 958-9. N Engl J Med. 2007 Mar 1;356(9):957; author reply 958-9. N Engl J Med. 2007 Mar 1;356(9):958; author reply 958-9. N Engl J Med. 2007 Mar 1;356(9):957-8; author reply 958-9. Nat Clin Pract Nephrol. 2007 May;3(5):244-5. doi: 10.1038/ncpneph0463. Semin Dial. 2007 May-Jun;20(3):277-9. doi: 10.1111/j.1525-139X.2007.00290.x. BACKGROUND: Anemia, a common complication of chronic kidney disease, usually develops as a consequence of erythropoietin deficiency. Recombinant human erythropoietin (epoetin alfa) is indicated for the correction of anemia associated with this condition. However, the optimal level of hemoglobin correction is not defined. METHODS: In this open-label trial, we studied 1432 patients with chronic kidney disease, 715 of whom were randomly assigned to receive a dose of epoetin alfa targeted to achieve a hemoglobin level of 13.5 g per deciliter and 717 of whom were assigned to receive a dose targeted to achieve a level of 11.3 g per deciliter. The median study duration was 16 months. The primary end point was a composite of death, myocardial infarction, hospitalization for congestive heart failure (without renal replacement therapy), and stroke. RESULTS: A total of 222 composite events occurred: 125 events in the high-hemoglobin group, as compared with 97 events in the low-hemoglobin group (hazard ratio, 1.34; 95% confidence interval, 1.03 to 1.74; P=0.03). There were 65 deaths (29.3%), 101 hospitalizations for congestive heart failure (45.5%), 25 myocardial infarctions (11.3%), and 23 strokes (10.4%). Seven patients (3.2%) were hospitalized for congestive heart failure and myocardial infarction combined, and one patient (0.5%) died after having a stroke. Improvements in the quality of life were similar in the two groups. More patients in the high-hemoglobin group had at least one serious adverse event. CONCLUSIONS: The use of a target hemoglobin level of 13.5 g per deciliter (as compared with 11.3 g per deciliter) was associated with increased risk and no incremental improvement in the quality of life. (ClinicalTrials.gov number, NCT00211120 [ClinicalTrials.gov].). Copyright 2006 Massachusetts Medical Society. DOI: 10.1056/NEJMoa065485 PMID: 17108343 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/17437743
1. Am J Cardiol. 2007 Apr 15;99(8):1137-42. doi: 10.1016/j.amjcard.2006.11.055. Epub 2007 Mar 7. Anemia as a risk factor for kidney function decline in individuals with heart failure. Bansal N(1), Tighiouart H, Weiner D, Griffith J, Vlagopoulos P, Salem D, Levin A, Sarnak MJ. Author information: (1)Division of Nephrology, Tufts-New England Medical Center, Boston, Massachusetts, USA. Chronic kidney disease (CKD), anemia, and declining kidney function are recognized as risk factors for adverse outcomes in patients with heart failure. This analysis was conducted to evaluate whether anemia is a risk factor for kidney function decrease in patients with heart failure. Data from the Studies of Left Ventricular Dysfunction (SOLVD), a randomized trial of enalapril versus placebo in patients with ejection fractions <or=35%, were analyzed. After randomization, creatinine measurements were taken at 2 weeks, 6 weeks, 4 months, and every 4 months thereafter. The glomerular filtration rate (GFR) was estimated using the Modification of Diet in Renal Disease Study (MDRD) equation, and GFR slope was calculated. "Rapid decrease" was defined as a decrease in the GFR of >or=6 ml/min/1.73 m(2)/year. Anemia was defined as baseline hematocrit <36%. Multivariate logistic regression weighted by the number of GFR assessments was used to test the relation between anemia and rapid decrease. We also evaluated whether CKD (baseline GFR </=60 ml/min/1.73 m(2)) modified the relation between anemia and rapid decrease. In the 6,360 subjects, the mean age was 59 years, 31% had CKD, and 6% had anemia. Median follow-up was 2 years. In multivariate analysis, anemia was associated with a 1.30 increased odds (95% confidence interval 1.18 to 1.45) of rapid decrease in GFR. In subjects with CKD, anemia was associated with a 1.71 increased odds (95% confidence interval 1.43 to 2.05) of rapid decrease, while in subjects without CKD, anemia was associated with a 1.16 increased odds (95% confidence interval 1.03 to 1.31) of rapid decrease (p for interaction <0.001). In conclusion, anemia is associated with a rapid decrease in kidney function in patients with heart failure, particularly in those with underlying CKD. DOI: 10.1016/j.amjcard.2006.11.055 PMID: 17437743 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/2010480
1. Hosp Pract (Off Ed). 1991 Apr 15;26(4):61-9. doi: 10.1080/21548331.1991.11704158. Recombinant erythropoietin and chronic renal failure. Lundin AP(1). Author information: (1)State University of New York, Brooklyn. The hormone deficiency that underlies anemia in chronic kidney failure can now be corrected. Along with the primary benefit of raising the hematocrit and reversing anemia, dramatic secondary benefits can be achieved. Exercise capacity, neuropsychiatric and sexual function, and overall quality of life are enhanced. Guidelines for management are discussed. DOI: 10.1080/21548331.1991.11704158 PMID: 2010480 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/2357306
1. Crit Care Nurs Clin North Am. 1990 Mar;2(1):101-13. Use of EPOGEN for treatment of anemia associated with chronic renal failure. Erlich L. The anemia of chronic renal failure is a major contributor to the symptoms that limit the quality of life and preclude rehabilitation. The primary cause of the anemia of chronic renal failure is an inability on the part of the impaired kidney to produce adequate amounts of erythropoietin. This hormone stimulates the bone marrow to produce red blood cells. EPOGEN (Epoietin alfa), a glycoprotein produced through recombinant DNA technology, corrects this anemia. Its contribution to the care of persons with chronic renal failure is explored. PMID: 2357306 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/22680636
1. Curr Med Chem. 2012;19(21):3502-7. doi: 10.2174/092986712801323261. Cardio-renal-anemia syndrome: a link between erythropoietin, dimethylarginine and homocysteine. Righetti M(1). Author information: (1)Nephrology & Dialysis Unit, Uboldo Hospital, Cernusco S/N (Milan), Italy. [email protected] Cardio-renal-anemia syndrome is a combination of heart failure, kidney failure, and anemia. Many advanced chronic kidney disease patients have both anemia and chronic heart failure. They have often hyperhomocysteinemia, high dimethylarginine values and low erythropoietin levels. Nephrologists treat advanced chronic kidney disease patients with erythropoiesis stimulating agents to improve anemia, renal and heart disease. Erythropoiesis stimulating agents, though considered essential to improve anemia in chronic kidney disease patients, have shown no significant protective effect on cardiovascular disease when used in large clinical trials targeting normal hemoglobin levels. It is possible that the high amounts of these drugs, given to reach normal hemoglobin values, may have counterbalanced the positive effect on endothelium obtained with low doses. Many studies have shown that erythropoietin improves endothelial function in animals with high dimethylarginine levels, lowering asymmetric dimethylarginine and increasing nitric oxide synthesis. Advanced chronic kidney disease patients have also high homocysteine levels which further reduce endothelial function by increasing asymmetric dimethylarginine. Homocysteine-lowering vitamin B treatment has been associated to a significant reduction of cardiovascular disease in advanced chronic kidney disease patients. Low doses of epoetin and B vitamins may improve cardiovascular morbidity by reducing asymmetric dimethylarginine and by increasing nitric oxide synthase activity. This review analyses the interaction between erythropoietin, dimethylarginine and homocysteine, and their role in cardio-renal-anemia syndrome. DOI: 10.2174/092986712801323261 PMID: 22680636 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/9375826
1. Nephron. 1997;77(3):315-8. doi: 10.1159/000190294. Relative contributions of body iron status and uremia severity to anemia in patients with advanced chronic renal failure. Ifudu O(1), Dawood M, Friedman EA. Author information: (1)Department of Medicine, State University of New York Health Science Center at Brooklyn, 11203, USA. Anemia in chronic renal failure is predominantly caused by diminished erythropoietin synthesis by diseased kidneys. While iron deficiency is often stated as a cause of anemia in chronic renal failure prior to end-stage renal disease, its relative contribution is debated. It is speculated that rather than frank 'iron deficiency', many patients with chronic renal failure may indeed have impaired utilization of iron. We analyzed 139 consecutive patients with chronic renal failure starting maintenance hemodialysis to determine the relationship between hematocrit, measures of renal function (blood urea nitrogen and serum creatinine concentration), and measures of iron availability (serum transferrin saturation, serum iron level and serum ferritin). The 139 study subjects (60 men, 79 women) comprised 116 blacks (83%), 15 hispanics (11%), and 8 whites (6%) of a mean age 56 +/- 15 years. Only 23 (17%) of 139 subjects had positive hemoccult stool test for blood. Their mean hematocrit was 24 +/- 4.5%, mean blood urea nitrogen concentration was 121 +/- 38, mean serum creatinine concentration was 12.6 +/- 5.2 mg/dl, mean serum transferrin saturation was 22 +/- 14%, mean serum ferritin level was 235 +/- 194 U/l, mean serum iron level was 55 +/- 40 U/l, and mean total iron binding capacity was 254 +/- 93%. Multiple regression analysis with hematocrit as the outcome variable, and blood urea nitrogen level, serum creatinine concentration, serum albumin concentration, serum transferrin saturation, and serum ferritin level as the independent variables, showed an inverse correlation between hematocrit and serum creatinine concentration (p = 0.002). We conclude that in patients with chronic renal failure starting uremia therapy, anemia does not correlate with any of the commonly measured indices of body iron stores. We infer that impaired utilization of iron may be a significant factor in the anemia of chronic renal failure. DOI: 10.1159/000190294 PMID: 9375826 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/18670732
1. Clin Exp Nephrol. 2009 Apr;13(2):101-106. doi: 10.1007/s10157-008-0074-1. Epub 2008 Aug 1. The correction of anemia in patients with the combination of chronic kidney disease and congestive heart failure may prevent progression of both conditions. Silverberg DS(1), Wexler D(2), Iaina A(3), Schwartz D(3). Author information: (1)Department of Nephrology, Tel Aviv Medical Center, Weizman 6, 64239, Tel Aviv, Israel. [email protected]. (2)Department of Cardiology and Heart Failure Unit, Tel Aviv Medical Center, Weizman 6, 64239, Tel Aviv, Israel. (3)Department of Nephrology, Tel Aviv Medical Center, Weizman 6, 64239, Tel Aviv, Israel. It has recently been recognized that many patients with congestive heart failure (CHF) are anemic. The anemia is very often associated with chronic kidney disease (CKD). The more severe the anemia the more severe the CHF, with higher mortality, morbidity, and hospitalization rate. The only way to prove that the anemia is itself a causative factor in the progression of both the CKD and the CHF is to correct it. In this paper we review the results of published papers and some preliminary reports about correction of this anemia in CHF. These studies frequently showed that erythropoietic stimulating agents (ESA) with oral or IV iron often resulted in improvement in left ventricular systolic and diastolic function, dilation, and hypertrophy, stabilization or improvement in renal function, reduced hospitalizations, diuretic dose, mitral regurgitation, pulmonary artery pressure, plasma volume, heart rate, serum brain natriuretic peptide levels, and the inflammatory markers C reactive protein and Interleukin 6, and an improvement in New York Heart Association class, exercise capacity, oxygen utilization during exercise, sleep apnea, caloric intake, depression, and quality of life. The activity of endothelial progenitor cells was also increased. Iron deficiency may also play an important role in the anemia, because significant improvement of cardiac, renal, and functional status in these anemic CKD-CHF has been seen after treatment with IV iron alone. Clearly more work is needed to clarify the relationship between anemia, CKD and CHF. DOI: 10.1007/s10157-008-0074-1 PMID: 18670732 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/12076240
1. Arch Intern Med. 2002 Jun 24;162(12):1401-8. doi: 10.1001/archinte.162.12.1401. Association of kidney function with anemia: the Third National Health and Nutrition Examination Survey (1988-1994). Astor BC(1), Muntner P, Levin A, Eustace JA, Coresh J. Author information: (1)Department of Epidemiology, The Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA. [email protected] BACKGROUND: Kidney failure is known to cause anemia, which is associated with a higher risk of cardiac failure and mortality. The impact of milder decreases in kidney function on hemoglobin levels and anemia in the US population, however, is unknown. METHODS: We analyzed a population-based sample of 15419 participants 20 years and older in the Third National Health and Nutrition Examination Survey, conducted from 1988 to 1994. RESULTS: Lower kidney function was associated with a lower hemoglobin level and a higher prevalence and severity of anemia below, but not above, an estimated glomerular filtration rate (GFR) of 60 mL/min per 1.73 m(2). Adjusted to the age of 60 years, the predicted median hemoglobin level among men (women) decreased from 14.9 (13.5) g/dL at an estimated GFR of 60 mL/min per 1.73 m(2) to 13.8 (12.2) g/dL at an estimated GFR of 30 mL/min per 1.73 m(2) and to 12.0 (10.3) g/dL at an estimated GFR of 15 mL/min per 1.73 m(2). The prevalence of anemia (hemoglobin level <12 g/dL in men and <11 g/dL in women) increased from 1% (95% confidence interval, 0.7%-2%) at an estimated GFR of 60 mL/min per 1.73 m(2) to 9% (95% confidence interval, 4%-19%) at an estimated GFR of 30 mL/min per 1.73 m(2) and to 33% (95% confidence interval, 11%-67%) at an estimated GFR of 15 mL/min per 1.73 m(2) among men and to 67% (95% confidence interval, 30%-90%) at an estimated GFR of 15 mL/min per 1.73 m(2) among women. An estimated GFR of 15 to 60 mL/min per 1.73 m(2) was present in 4% of the entire population and in 17% of the individuals with anemia. CONCLUSION: Below an estimated GFR of 60 mL/min per 1.73 m(2), lower kidney function is strongly associated with a higher prevalence of anemia among the US adult population. DOI: 10.1001/archinte.162.12.1401 PMID: 12076240 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/22536082
1. Int J Nephrol Renovasc Dis. 2012;5:53-60. doi: 10.2147/IJNRD.S23447. Epub 2012 Mar 30. Methoxy polyethylene glycol-epoetin beta for anemia with chronic kidney disease. Ohashi N(1), Sakao Y, Yasuda H, Kato A, Fujigaki Y. Author information: (1)Internal Medicine 1, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan. Chronic kidney disease (CKD) is a risk factor for end-stage renal failure and cardiovascular events. In patients with CKD, anemia is often caused by decreased erythropoietin production relative to hemoglobin levels. As correction of anemia is associated with improved cardiac and renal function and quality of life, erythropoiesis-stimulating agents (ESAs) are standard therapy for anemia in CKD patients. However, traditional ESAs such as epoetin or darbepoetin have short half-lives and require frequent administration, dose changes, and close monitoring of hemoglobin concentration to maintain target hemoglobin levels. Methoxy polyethylene glycol-epoetin beta (MPG-EPO) is the only ESA that is generated by chemical modification of glycosylated erythropoietin through the integration of one specific, long, linear chain of polyethylene glycol. This ESA induces continuous erythropoietin receptor activation and has a long half-life (approximately 130 hours). Subcutaneous or intravenous administration of MPG-EPO once every 2 weeks or monthly achieved a high hemoglobin response rate in patients with anemia associated with CKD, regardless of whether the patient was undergoing dialysis. According to data from an observational time and motion study, MPG-EPO maintains hemoglobin levels when the same dose is administered, however infrequently. This suggests that compared with the use of traditional ESAs, administration of MPG-EPO reduces the overall time and cost associated with the management of anemia in CKD patients undergoing dialysis. MPG-EPO is generally well tolerated and most adverse events are of mild to moderate severity. The most commonly reported adverse effects are hypertension, nasopharyngitis, and diarrhea. Subcutaneous injection of MPG-EPO is significantly less painful than subcutaneous injection of darbepoetin. In conclusion, MPG-EPO is as effective and safe as traditional ESAs in managing renal anemia, irrespective of whether the patient is undergoing dialysis. DOI: 10.2147/IJNRD.S23447 PMCID: PMC3333806 PMID: 22536082
http://www.ncbi.nlm.nih.gov/pubmed/19833421
1. Blood Rev. 2010 Jan;24(1):39-47. doi: 10.1016/j.blre.2009.09.001. Epub 2009 Oct 14. Anemia in renal disease: diagnosis and management. Lankhorst CE(1), Wish JB. Author information: (1)Division of Nephrology, University Hospitals Case Medical Center, 11100 Euclid Ave, Cleveland, OH 44106, USA. [email protected] Chronic kidney disease (CKD) is a widespread health problem in the world and anemia is a common complication. Anemia conveys significant risk for cardiovascular disease, faster progression of renal failure and decreased quality of life. Patients with CKD can have anemia for many reasons, including but not invariably their renal insufficiency. These patients require a thorough evaluation to identify and correct causes of anemia other than erythropoietin deficiency. The mainstay of treatment of anemia secondary to CKD has become erythropoiesis-stimulating agents (ESAs). The use of ESAs does carry risks and these agents need to be used judiciously. Iron deficiency often co-exists in this population and must be evaluated and treated. Correction of iron deficiency can improve anemia and reduce ESA requirements. Partial, but not complete, correction of anemia is associated with improved outcomes in patients with CKD. Copyright 2009 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.blre.2009.09.001 PMID: 19833421 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32899941
1. Cells. 2020 Sep 6;9(9):2039. doi: 10.3390/cells9092039. Uremic Toxins Affect Erythropoiesis during the Course of Chronic Kidney Disease: A Review. Hamza E(1), Metzinger L(1), Metzinger-Le Meuth V(1)(2). Author information: (1)HEMATIM UR 4666, C.U.R.S, University of Picardie Jules Verne, CEDEX 1, 80025 Amiens, France. (2)INSERM UMRS 1148, Laboratory for Vascular Translational Science (LVTS), UFR SMBH, University of Paris Nord, 93000 Bobigny, France. Chronic kidney disease (CKD) is a global health problem characterized by progressive kidney failure due to uremic toxicity and the complications that arise from it. Anemia consecutive to CKD is one of its most common complications affecting nearly all patients with end-stage renal disease. Anemia is a potential cause of cardiovascular disease, faster deterioration of renal failure and mortality. Erythropoietin (produced by the kidney) and iron (provided from recycled senescent red cells) deficiencies are the main reasons that contribute to CKD-associated anemia. Indeed, accumulation of uremic toxins in blood impairs erythropoietin synthesis, compromising the growth and differentiation of red blood cells in the bone marrow, leading to a subsequent impairment of erythropoiesis. In this review, we mainly focus on the most representative uremic toxins and their effects on the molecular mechanisms underlying anemia of CKD that have been studied so far. Understanding molecular mechanisms leading to anemia due to uremic toxins could lead to the development of new treatments that will specifically target the pathophysiologic processes of anemia consecutive to CKD, such as the newly marketed erythropoiesis-stimulating agents. DOI: 10.3390/cells9092039 PMCID: PMC7565991 PMID: 32899941 [Indexed for MEDLINE] Conflict of interest statement: The authors declare no conflict of interest.
http://www.ncbi.nlm.nih.gov/pubmed/29151105
1. Kidney Blood Press Res. 2017;42(4):749-760. doi: 10.1159/000484215. Epub 2017 Oct 23. Eryptosis - the Neglected Cause of Anemia in End Stage Renal Disease. Lang F(1)(2), Bissinger R(3), Abed M(1), Artunc F(4)(5)(6). Author information: (1)Department of Physiology I, University of Tübingen, Tübingen, Germany. (2)Department of Molecular Medicine II, Medical Faculty, Heinrich Heine University, Düsseldorf, Germany. (3)Department of Internal Medicine III, University of Tübingen, Tübingen, Germany. (4)Department of Internal Medicine IV, Division of Endocrinology, Diabetology, Vascular Disease, Nephrology and Clinical Chemistry, University Hospital Tübingen, Tübingen, Germany. (5)Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University Tübingen, Tübingen, Germany. (6)German Center for Diabetes Research (DZD) at the University Tübingen, Tübingen, Germany. End stage renal disease (ESRD) invariably leads to anemia which has been mainly attributed to compromised release of erythropoietin from the defective kidneys with subsequent impairment of erythropoiesis. However, erythropoietin replacement only partially reverses anemia pointing to the involvement of additional mechanisms. As shown more recently, anemia of ESRD is indeed in large part a result of accelerated erythrocyte loss due to suicidal erythrocyte death or eryptosis, characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine translocation to the cell surface. Phosphatidylserine exposing erythrocytes are bound to and engulfed by macrophages and are thus rapidly cleared from circulating blood. If the loss of erythrocytes cannot be fully compensated by enhanced erythropoiesis, stimulation of eryptosis leads to anemia. Eryptotic erythrocytes may further adhere to the vascular wall and thus impair microcirculation. Stimulators of eryptosis include complement, hyperosmotic shock, energy depletion, oxidative stress, and a wide variety of xenobiotics. Signaling involved in the stimulation of eryptosis includes increase of cytosolic Ca2+ activity, ceramide, caspases, calpain, p38 kinase, protein kinase C, Janus-activated kinase 3, casein kinase 1α, and cyclin-dependent kinase 4. Eryptosis is inhibited by AMP-activated kinase, p21-activated kinase 2, cGMP-dependent protein kinase, mitogen- and stress-activated kinase MSK1/2, and some illdefined tyrosine kinases. In ESRD eryptosis is stimulated at least in part by a plasma component, as it is triggered by exposure of erythrocytes from healthy individuals to plasma from ESRD patients. Several eryptosis-stimulating uremic toxins have been identified, such as vanadate, acrolein, methylglyoxal, indoxyl sulfate, indole-3-acetic acid and phosphate. Attempts to fully reverse anemia in ESRD with excessive stimulation of erythropoiesis enhances the number of circulating suicidal erythrocytes and bears the risk of interference with micocirculation, At least in theory, anemia in ESRD could preferably be treated with replacement of erythropoietin and additional inhibition of eryptosis thus avoiding eryptosis-induced impairment of microcirculation. A variety of eryptosis inhibitors have been identified, their efficacy in ESRD remains, however, to be shown. © 2017 The Author(s). Published by S. Karger AG, Basel. DOI: 10.1159/000484215 PMID: 29151105 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/20443766
1. Cardiovasc Hematol Agents Med Chem. 2010 Jul;8(3):164-72. doi: 10.2174/187152510791698398. Erythropoiesis stimulating agents and anaemia of end-stage renal disease. Schmid H(1), Schiffl H. Author information: (1)KfH Nierenzentrum Muenchen-Laim, Munich, Germany. Chronic kidney disease (CKD) is a widespread health problem in the world and anaemia of renal origin is a common problem. Anaemia associated with CKD covers significant risk for faster progression of chronic renal failure, decreased quality of life, and clinical manifestation of cardiovascular disease. The mainstay of anaemia treatment secondary to end-stage renal disease (ESRD) has become erythropoiesis stimulating agents (ESAs). More than 90 % of ESRD patients maintained on dialysis respond to traditional recombinant human erythropoietin (rHU EPO) or to EPO analogues, also called "biosimilars". Iron deficiency often co-exists in dialysis patients and must be evaluated and treated to reduce ESA requirements. Partial, but not complete correction of renal anaemia is associated with improved outcomes in patients with CKD. The use of ESAs does carry risks such as hypertension, pure red cell aplasia, or cancer, and these agents need to be used judiciously. DOI: 10.2174/187152510791698398 PMID: 20443766 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/26948278
1. Proteomics Clin Appl. 2016 Aug;10(8):778-90. doi: 10.1002/prca.201500127. Epub 2016 Mar 29. Red blood cell abnormalities and the pathogenesis of anemia in end-stage renal disease. Georgatzakou HT(1), Antonelou MH(1), Papassideri IS(1), Kriebardis AG(2). Author information: (1)Department of Cell Biology and Biophysics, Faculty of Biology, National and Kapodistrian University of Athens, Greece. (2)Department of Medical Laboratories, Faculty of Health and Caring Professions, Technological and Educational Institute of Athens, Greece. Anemia is the most common hematologic complication in end-stage renal disease (ESRD). It is ascribed to decreased erythropoietin production, shortened red blood cell (RBC) lifespan, and inflammation. Uremic toxins severely affect RBC lifespan; however, the implicated molecular pathways are poorly understood. Moreover, current management of anemia in ESRD is controversial due to the "anemia paradox" phenomenon, which underlines the need for a more individualized approach to therapy. RBCs imprint the adverse effects of uremic, inflammatory, and oxidative stresses in a context of structural and functional deterioration that is associated with RBC removal signaling and morbidity risk. RBCs circulate in hostile plasma by raising elegant homeostatic defenses. Variability in primary defect, co-morbidity, and therapeutic approaches add complexity to the pathophysiological background of the anemic ESRD patient. Several blood components have been suggested as biomarkers of anemia-related morbidity and mortality risk in ESRD. However, a holistic view of blood cell and plasma modifications through integrated omics approaches and high-throughput studies might assist the development of new diagnostic tests and therapies that will target the underlying pathophysiologic processes of ESRD anemia. © 2016 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim. DOI: 10.1002/prca.201500127 PMID: 26948278 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/9241712
1. Semin Nephrol. 1997 Jul;17(4):257-69. Anemia, hypertension, and myocardial dysfunction in end-stage renal disease. Venkatesan J(1), Henrich WL. Author information: (1)Department of Medicine, Medical College of Ohio, Toledo 43699-0008, USA. Cardiovascular disease remains the major cause of mortality in patients with end stage renal disease (ESRD). The pathophysiology of cardiac dysfunction in ESRD is complex and not fully understood. However, it appears that the two major determinants of left ventricular (LV) hypertrophy and dysfunction are anemia and hypertension, both of which are very common in ESRD patients. Early and aggressive correction of anemia and hypertension may have a significant impact on cardiac disease in ESRD patients. This article presents a discussion on the management of anemia and hypertension, and the current information available on the pathogenesis and management of LV dysfunction in ESRD. PMID: 9241712 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/7034633
1. Ann Clin Lab Sci. 1981 Nov-Dec;11(6):484-7. Anemia of chronic renal failure. Sexauer CL, Matson JR. Anemia is one of the most characteristic and visable manifestations of chronic renal failure. Investigators in the past decade have provided a better understanding of this anemia. The etiology of the anemia of chronic renal failure has three facets: first is reduced erythropoietin production by damaged kidneys; second is the presence of inhibitors to red blood cell (RBC) production in uremic serum; and third is red blood cell hemolysis. Unfortunately, transfusion therapy with its expense and risk of transmissable viral disease remains the mainstay of management for symptomatic anemia. Other modalities include dialysis, androgens, histidine supplementation, and erythropoietin replacement. PMID: 7034633 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31994917
1. Arch Physiol Biochem. 2022 Jun;128(3):703-707. doi: 10.1080/13813455.2020.1720742. Epub 2020 Jan 29. Association between neutrophil gelatinase-associated lipocalin (NGAL) and iron profile in chronic renal disease. Aghsaeifard Z(1)(2), Alizadeh R(3), Bagheri N(4). Author information: (1)Department of Internal Medicine, School of Medicine, Sina Hospital Tehran University of Medical Sciences, Tehran, Iran. (2)Urology Research Center, Tehran University of Medical Sciences, Tehran, Iran. (3)Department of Anesthesiology and Intensive Care, AJA University of Medical Sciences, Tehran, Iran. (4)Department of Adult Nephrology, School of Medicine, Ayatollah Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran. NGAL, also known as lipocalin 2, is a stress protein located on the cell surface that is known for its involvement in iron transport. This study is aimed to evaluate the correlation between the iron profile and NGAL concentration in serum among chronic kidney disease patients under dialysis in order to find its diagnostic value with regards to iron deficiency anaemia (IDA). 47 patients under chronic haemodialysis in end-stage renal disease (ESRD) and 15 healthy controls were evaluated to determine the correlation between serum NGAL concentration and IDA characteristics. Our results recorded a significant correlation between IDA (TSAT < 20%) and NGAL serum concentration with a Spearman's coefficient of 0.314. Serum NGAL was also significantly related to serum ferritin, TIBC, uric acid, creatinine and blood sugar whereas, an inverse relationship with albumin, total cholesterol and LDL. Our study reports a positive correlation between IDA and serum NGAL levels in CKD patients. DOI: 10.1080/13813455.2020.1720742 PMID: 31994917 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/26785833
1. Eur J Hum Genet. 2016 Aug;24(9):1255-61. doi: 10.1038/ejhg.2015.283. Epub 2016 Jan 20. Online genetic counseling from the providers' perspective: counselors' evaluations and a time and cost analysis. Otten E(1), Birnie E(1), Ranchor AV(2), van Langen IM(1). Author information: (1)Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands. (2)Department of Health Psychology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands. Telemedicine applications are increasingly being introduced in patient care in various disciplines, including clinical genetics, mainly to increase access to care and to reduce time and costs for patients and professionals. Most telegenetics reports describe applications in large geographical areas, showing positive patients' and professionals' satisfaction. One economic analysis published thus far reported lower costs than in-person care. We hypothesized that telegenetics can also be beneficial from the professional's view in relatively small geographical areas. We performed a pilot study in the Northern Netherlands of 51 home-based online counseling sessions for cardiogenetic and oncogenetic cascade screening, and urgent prenatal counseling. Previously, we showed patient satisfaction, anxiety, and perceived control of online counseling to be comparable to in-person counseling. This study focuses on expectations, satisfaction, and practical evaluations of the involved counselors, and the impact in terms of time and costs. Most counselors expected disadvantages of online counseling for themselves and their patients, mainly concerning insufficient non-verbal communication; few expected advantages for themselves. Afterwards, counselors additionally raised the disadvantage of insufficient verbal communication, and reported frequent technical problems. Their overall mean telemedicine satisfaction itemscore was 3.38 before, and 2.95 afterwards, being afterwards slightly below the minimum level we set for a satisfactory result. We estimated reduced time and costs by online counseling with about 8% and 10-12%, respectively. We showed online genetic counseling to be effective, feasible and cost-efficient, but technical improvements are needed to increase counselors' satisfaction. DOI: 10.1038/ejhg.2015.283 PMCID: PMC4989197 PMID: 26785833 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35723111
1. J Neuromuscul Dis. 2022;9(4):463-476. doi: 10.3233/JND-210776. Comparing Deflazacort and Prednisone in Duchenne Muscular Dystrophy. Biggar WD(1), Skalsky A(2), McDonald CM(3). Author information: (1)University of Toronto, 15583 22nd Side Road, Georgetown, Ontario, Canada. (2)University of California San Diego, Rady Children's Hospital San Diego, MC, San Diego, CA, USA. (3)University of California Davis Health, Departments of Physical Medicine & Rehabilitation and Pediatrics, Lawrence J. Ellison Ambulatory Care Center, Sacramento, CA, USA. Deflazacort and prednisone/prednisolone are the current standard of care for patients with Duchenne muscular dystrophy (DMD) based on evidence that they improve muscle strength, improve timed motor function, delay loss of ambulation, improve pulmonary function, reduce the need for scoliosis surgery, delay onset of cardiomyopathy, and increase survival. Both have been used off-label for many years (choice dependent on patient preference, cost, and geographic location) before FDA approval of deflazacort for DMD in 2017. In this review, we compare deflazacort and prednisone/prednisolone in terms of their key pharmacological features, relative efficacy, and safety profiles in patients with DMD. Differentiating features include lipid solubility, pharmacokinetics, changes in gene expression profiles, affinity for the mineralocorticoid receptor, and impact on glucose metabolism. Evidence from randomized clinical trials, prospective studies, meta-analyses, and post-hoc analyses suggests that patients receiving deflazacort experience similar or slower rates of functional decline compared with those receiving prednisone/prednisolone. Regarding side effects, weight gain and behavior side effects appear to be greater with prednisone/prednisolone than with deflazacort, whereas bone health, growth parameters, and cataracts appear worse with deflazacort. DOI: 10.3233/JND-210776 PMCID: PMC9398085 PMID: 35723111 [Indexed for MEDLINE] Conflict of interest statement: W. Douglas Biggar, MD has served as a consultant for PTC Therapeutics and Marathon. Andrew Skalsky, MD has no conflicts of interest to report. Craig M. McDonald, MD has served as a consultant for clinical trials for PTC Therapeutics, and outside the submitted work with Astellas Pharma, Avidity Biosciences, Capricor Therapeutics, Catabasis Pharmaceuticals, Edgewise Therapeutics, Entrada Therapeutics, Epirium Bio (formerly Cardero Therapeutics), FibroGen, Italfarmaco, Pfizer, PTC Therapeutics, Roche, Santhera Pharmaceuticals and Sarepta Therapeutics. He has received research support for clinical trials from Capricor Therapeutics, Catabasis Pharmaceuticals, Italfarmaco, Pfizer, PTC Therapeutics, Santhera Pharmaceuticals and Sarepta Therapeutics. He serves on external advisory boards related to Duchenne muscular dystrophy for PTC Therapeutics, Edgewise Therapeutics, Eli Lilly, Sarepta Therapeutics, Santhera Pharmaceuticals, and Capricor Therapeutics. The authors received no compensation for writing this article.
http://www.ncbi.nlm.nih.gov/pubmed/1822804
1. Neuromuscul Disord. 1991;1(4):261-6. doi: 10.1016/0960-8966(91)90099-e. Steroids in Duchenne muscular dystrophy--deflazacort trial. Mesa LE(1), Dubrovsky AL, Corderi J, Marco P, Flores D. Author information: (1)Sección de Enfermedades Neuromusculares, Hospital Frances, Buenos Aires, Argentina. We conducted a double blind controlled trial in 28 Duchenne muscular dystrophy (DMD) patients with Deflazacort (DF), an oxazoline derivative of prednisolone which reduces its side-effects. Myometric muscle strength measurements, Scott Score and timed tests showed statistically significant improvement for the treated group (P less than 0.05). Side-effects after 9 months of treatment included mild cushingoid appearance in four patients (28%) and moderate in only one (7%), increased appetite in seven (50%), increased body hair in four (28%), irritability and hyperactivity in three (21%). Increased body weight was not prominent and was controlled with dietary measures. No patient had to be withdrawn from medication. More research and long-term follow-up are needed in order to establish the mechanism of improvement and the consequences of long-term steroid administration in DMD. In this regard DF appears as an alternative to prednisone preserving its benefits but with fewer side-effects. DOI: 10.1016/0960-8966(91)90099-e PMID: 1822804 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/33104035
1. J Neuromuscul Dis. 2021;8(1):39-52. doi: 10.3233/JND-200556. Mechanisms and Clinical Applications of Glucocorticoid Steroids in Muscular Dystrophy. Quattrocelli M(1)(2), Zelikovich AS(1), Salamone IM(1), Fischer JA(1), McNally EM(1). Author information: (1)Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA. (2)Molecular Cardiovascular Biology Division, Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA. Glucocorticoid steroids are widely used as immunomodulatory agents in acute and chronic conditions. Glucocorticoid steroids such as prednisone and deflazacort are recommended for treating Duchenne Muscular Dystrophy where their use prolongs ambulation and life expectancy. Despite this benefit, glucocorticoid use in Duchenne Muscular Dystrophy is also associated with significant adverse consequences including adrenal suppression, growth impairment, poor bone health and metabolic syndrome. For other forms of muscular dystrophy like the limb girdle dystrophies, glucocorticoids are not typically used. Here we review the experimental evidence supporting multiple mechanisms of glucocorticoid action in dystrophic muscle including their role in dampening inflammation and myofiber injury. We also discuss alternative dosing strategies as well as novel steroid agents that are in development and testing, with the goal to reduce adverse consequences of prolonged glucocorticoid exposure while maximizing beneficial outcomes. DOI: 10.3233/JND-200556 PMCID: PMC7902991 PMID: 33104035 [Indexed for MEDLINE] Conflict of interest statement: MQ and EMM are listed as co-inventors on a patent application related to intermittent glucocorticoid use filed by Northwestern University.
http://www.ncbi.nlm.nih.gov/pubmed/35381069
1. JAMA. 2022 Apr 19;327(15):1456-1468. doi: 10.1001/jama.2022.4315. Effect of Different Corticosteroid Dosing Regimens on Clinical Outcomes in Boys With Duchenne Muscular Dystrophy: A Randomized Clinical Trial. Guglieri M(1), Bushby K(1), McDermott MP(2)(3), Hart KA(2), Tawil R(2), Martens WB(2), Herr BE(2), McColl E(4), Speed C(4)(5), Wilkinson J(4), Kirschner J(6)(7), King WM(8), Eagle M(1), Brown MW(2), Willis T(9), Griggs RC(2); FOR-DMD Investigators of the Muscle Study Group; Straub V(1), van Ruiten H(1), Childs AM(10), Ciafaloni E(2), Shieh PB(11), Spinty S(12), Maggi L(13), Baranello G(13)(14), Butterfield RJ(15), Horrocks IA(16), Roper H(17), Alhaswani Z(17), Flanigan KM(18), Kuntz NL(19), Manzur A(20), Darras BT(21), Kang PB(21)(22), Morrison L(23), Krzesniak-Swinarska M(23), Mah JK(24), Mongini TE(25), Ricci F(25), von der Hagen M(26), Finkel RS(27)(28), O'Reardon K(27), Wicklund M(29)(30), Kumar A(29), McDonald CM(31), Han JJ(31), Joyce N(31), Henricson EK(31), Schara-Schmidt U(32), Gangfuss A(32), Wilichowski E(33), Barohn RJ(34), Statland JM(35), Campbell C(36), Vita G(37), Vita GL(38), Howard JF Jr(39), Hughes I(40), McMillan HJ(41), Pegoraro E(42), Bello L(42), Burnette WB(43), Thangarajh M(44), Chang T(45). Author information: (1)John Walton Muscular Dystrophy Research Centre, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, England. (2)Department of Neurology, University of Rochester Medical Center, Rochester, New York. (3)Department of Biostatistics and Computational Biology, University of Rochester Medical Center, Rochester, New York. (4)Newcastle University, Newcastle upon Tyne, England. (5)NIHR Clinical Research Network North East and North Cumbria, Newcastle upon Tyne, England. (6)Department of Neuropediatrics and Muscle Disorders, Faculty of Medicine, University Hospital Freiburg, Freiburg, Germany. (7)Department of Neuropediatrics, University Hospital Bonn, Bonn, Germany. (8)Ohio State University, Columbus. (9)Robert Jones and Agnes Hunt Orthopaedic Hospital, NHS Foundation Trust, Oswestry, England. (10)Leeds General Infirmary, Leeds, England. (11)David Geffen School of Medicine, University of California, Los Angeles. (12)Alderhey Children's Hospital NHS Foundation Trust, Liverpool, England. (13)Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy. (14)Dubowitz Neuromuscular Centre, UCL NIHR GOSH Biomedical Research Centre, UCL Great Ormond Street Institute of Child Health, London, England. (15)Department of Pediatric Neurology, University of Utah, Salt Lake City. (16)Glasgow Paediatric Neuromuscular Research Centre, Greater Glasgow and Clyde NHS Yorkhill Hospital, Glasgow, Scotland. (17)University Hospitals Birmingham NHS Foundation Trust, Birmingham, England. (18)Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, Ohio. (19)Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois. (20)Great Ormond Street Hospital, London, England. (21)Department of Neurology, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts. (22)Paul and Sheila Wellstone Muscular Dystrophy Center and Department of Neurology, University of Minnesota Medical School, Minneapolis. (23)Health Sciences Center, University of New Mexico, Albuquerque. (24)Cumming School of Medicine, University of Calgary and Alberta Children's Hospital Research Institute, Calgary, Canada. (25)Neuromuscular Center, AOU Città della Salute e della Scienza, University of Turin, Turin, Italy. (26)Abteilung Neuropädiatrie, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany. (27)Nemours Children's Hospital, Orlando, Florida. (28)Center for Experimental Neurotherapeutics, St Jude Children's Research Hospital, Memphis, Tennessee. (29)Penn State Health Milton S. Hershey Medical Center, Hershey, Pennsylvania. (30)Department of Neurology, School of Medicine, University of Colorado, Aurora. (31)Department of Physical Medicine and Rehabilitation and Department of Pediatrics, University of California-Davis, Sacramento. (32)Clinic for Pediatrics I, Pediatric Neurology, University Hospital Essen, Essen, Germany. (33)Department of Pediatrics and Adolescent Medicine, Division of Pediatric Neurology, University Medical Center Göttingen, Göttingen, Germany. (34)University of Missouri, Columbia. (35)University of Kansas Medical Center, Kansas City. (36)Departments of Pediatrics, Clinical Neurological Sciences, and Epidemiology, University of Western Ontario, London, Canada. (37)ERN Neuromuscular Unit, Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy. (38)Unit of Neurology, IRCCS Centro Neurolesi Bonino-Pulejo, Messina, Italy. (39)School of Medicine, University of North Carolina, Chapel Hill. (40)Royal Manchester Children's Hospital, Manchester, England. (41)Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, Canada. (42)ERN Neuromuscular Unit, Department of Neuroscience, University of Padova, Padua, Italy. (43)Vanderbilt University Medical Center, Nashville, Tennessee. (44)Department of Neurology, Virginia Commonwealth University, Richmond. (45)Children's National Hospital, George Washington University, Washington, DC. IMPORTANCE: Corticosteroids improve strength and function in boys with Duchenne muscular dystrophy. However, there is uncertainty regarding the optimum regimen and dosage. OBJECTIVE: To compare efficacy and adverse effects of the 3 most frequently prescribed corticosteroid regimens in boys with Duchenne muscular dystrophy. DESIGN, SETTING, AND PARTICIPANTS: Double-blind, parallel-group randomized clinical trial including 196 boys aged 4 to 7 years with Duchenne muscular dystrophy who had not previously been treated with corticosteroids; enrollment occurred between January 30, 2013, and September 17, 2016, at 32 clinic sites in 5 countries. The boys were assessed for 3 years (last participant visit on October 16, 2019). INTERVENTIONS: Participants were randomized to daily prednisone (0.75 mg/kg) (n = 65), daily deflazacort (0.90 mg/kg) (n = 65), or intermittent prednisone (0.75 mg/kg for 10 days on and then 10 days off) (n = 66). MAIN OUTCOMES AND MEASURES: The global primary outcome comprised 3 end points: rise from the floor velocity (in rise/seconds), forced vital capacity (in liters), and participant or parent global satisfaction with treatment measured by the Treatment Satisfaction Questionnaire for Medication (TSQM; score range, 0 to 100), each averaged across all study visits after baseline. Pairwise group comparisons used a Bonferroni-adjusted significance level of .017. RESULTS: Among the 196 boys randomized (mean age, 5.8 years [SD, 1.0 years]), 164 (84%) completed the trial. Both daily prednisone and daily deflazacort were more effective than intermittent prednisone for the primary outcome (P < .001 for daily prednisone vs intermittent prednisone using a global test; P = .017 for daily deflazacort vs intermittent prednisone using a global test) and the daily regimens did not differ significantly (P = .38 for daily prednisone vs daily deflazacort using a global test). The between-group differences were principally attributable to rise from the floor velocity (0.06 rise/s [98.3% CI, 0.03 to 0.08 rise/s] for daily prednisone vs intermittent prednisone [P = .003]; 0.06 rise/s [98.3% CI, 0.03 to 0.09 rise/s] for daily deflazacort vs intermittent prednisone [P = .017]; and -0.004 rise/s [98.3% CI, -0.03 to 0.02 rise/s] for daily prednisone vs daily deflazacort [P = .75]). The pairwise comparisons for forced vital capacity and TSQM global satisfaction subscale score were not statistically significant. The most common adverse events were abnormal behavior (22 [34%] in the daily prednisone group, 25 [38%] in the daily deflazacort group, and 24 [36%] in the intermittent prednisone group), upper respiratory tract infection (24 [37%], 19 [29%], and 24 [36%], respectively), and vomiting (19 [29%], 17 [26%], and 15 [23%]). CONCLUSIONS AND RELEVANCE: Among patients with Duchenne muscular dystrophy, treatment with daily prednisone or daily deflazacort, compared with intermittent prednisone alternating 10 days on and 10 days off, resulted in significant improvement over 3 years in a composite outcome comprising measures of motor function, pulmonary function, and satisfaction with treatment; there was no significant difference between the 2 daily corticosteroid regimens. The findings support the use of a daily corticosteroid regimen over the intermittent prednisone regimen tested in this study as initial treatment for boys with Duchenne muscular dystrophy. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT01603407. DOI: 10.1001/jama.2022.4315 PMCID: PMC8984930 PMID: 35381069 [Indexed for MEDLINE] Conflict of interest statement: Conflict of Interest Disclosures: Dr Guglieri reported receiving grants from Duchenne UK, the European Union’s Horizon 2020 program for the Vision-DMD study (in collaboration with ReveraGen BioPharma Inc), and Sarepta Therapeutics; serving as a consultant to Dyne Therapeutics Inc, Pfizer, and NS Pharma Inc; receiving personal fees from Sarepta Therapeutics; and receiving nonfinancial support from Italfarmaco, Pfizer, ReveraGen BioPharma Inc, and Santhera Pharmaceuticals. Dr McDermott reported receiving grant support from PTC Therapeutics and receiving personal fees from Fulcrum Therapeutics, NeuroDerm Ltd, AstraZeneca, Eli Lilly, Catabasis Pharmaceuticals, Vaccinex Inc, Cynapsus Therapeutics, Neurocrine Biosciences, Voyager Therapeutics, Prilenia Therapeutics, ReveraGen BioPharma Inc, and NS Pharma Inc. Ms Hart reported receiving personal fees from Sarepta Therapeutics, Santhera Pharmaceuticals, and PTC Therapeutics. Dr McColl reported receiving grants from the National Institute for Health Research. Dr Kirschner reported receiving personal fees from PTC Therapeutics, Pfizer, Roche, and Sarepta Therapeutics. Dr Eagle reported receiving personal fees from ATOM International Ltd. Dr Willis reported receiving personal fees from Newcastle University, Novartis, Biogen Inc, Sarepta, Santhera, PTC Therapeutics, and Sanofi Genzyme. Dr Straub reported receiving grants from Sarepta Therapeutics and Sanofi Genzyme and serving as a consultant to Sarepta Therapeutics, Edgewise Therapeutics, Sanofi Genzyme, Vertex Pharmaceuticals, Biogen Inc, Roche, Novartis Gene Therapies, Astellas Gene Therapies, Wave Therapeutics, Kate Therapeutics Inc, and Pfizer. Dr Childs reported receiving personal fees from PTC Therapeutics, Sarepta Therapeutics, FibroGen Inc, the University of Basel, and ReveraGen BioPharma Inc. Dr Ciafaloni reported receiving personal fees from Viela Bio, AveXis Inc (now Novartis Gene Therapies), Biogen Inc, Medscape, Pfizer, PTC Therapeutics, Sarepta Therapeutics, Ra Pharmaceuticals, Wave Therapeutics, and Strongbridge Biopharma; receiving grants from the US Centers for Disease Control and Prevention, Cure SMA, the Muscular Dystrophy Association, the National Institutes of Health, Orphazyme, the Patient-Centered Outcomes Research Institute, Parent Project Muscular Dystrophy, PTC Therapeutics, Santhera Pharmaceuticals, Sarepta Therapeutics, and the US Food and Drug Administration; receiving royalties from Oxford University Press; and receiving compensation from MedLink for editorial duties. Dr Shieh reported receiving grants from Catalyst Pharmaceuticals, Sarepta Therapeutics, PTC Therapeutics, Pfizer, Sanofi, AveXis (now Novartis Gene Therapies), Biogen Inc, Santhera Pharmaceuticals, Fulcrum Therapeutics Inc, Astellas Gene Therapies, Solid Biosciences, ReveraGen BioPharma Inc, AMO Pharma, and Zogenix Inc and receiving personal fees from Sarepta Therapeutics, Ra Pharmaceuticals, Argenx, PTC Therapeutics, Pfizer, CSL Behring, Genentech, AveXis, Grifols, Alexion Pharmaceuticals, and Biogen Inc. Mr Spinty reported receiving personal fees from Roche, Sarepta Therapeutics, Pfizer, and Biogen Inc and receiving grants from PTC Therapeutics. Dr Maggi reported receiving personal fees from Biogen Inc, Sanofi-Genzyme, and Roche. Dr Baranello reported receiving personal fees from PTC Therapeutics, Sarepta Therapeutics, ReveraGen BioPharma Inc, NS Pharma Inc, Pfizer, and Roche. Dr Butterfield reported receiving personal fees from AavantiBio, Scholar Rock Inc, AveXis (now Novartis Gene Therapies), Pfizer, PTC Therapeutics, Biogen Inc, and Sarepta Therapeutics. Dr Roper reported reported receiving personal fees from GlaxoSmithKline, Prosensa, BioMarin Pharmaceutical Inc, and Biogen Inc and receiving grants from Summit Therapeutics and Roche. Dr Flanigan reported receiving grants and royalty payments from Astellas Gene Therapies and receiving grants from Sarepta Therapeutics. Dr Kuntz reported receiving personal fees from Sarepta Therapeutics, Astellas Gene Therapies, Novartis, and Genentech. Dr Manzur reported receiving grants from Muscular Dystrophy UK. Dr Darras reported receiving personal fees from Amicus Inc, Audentes Therapeutics (now Astellas Gene Therapies), AveXis (now Novartis Gene Therapies), Biogen Inc, Pfizer, Vertex, Roche, and Genentech; receiving research support from the National Institute of Neurological Disorders and Stroke, the Slaney Family Fund, the Spinal Muscular Atrophy Foundation, Cure SMA, and Working on Walking Fund; receiving grants from Ionis Pharmaceuticals Inc, Biogen Inc, AveXis, Sarepta Therapeutics, Novartis, PTC Therapeutics, Roche, Scholar Rock Inc, and FibroGen Inc; and receiving royalties for books and online publications from Elsevier and UpToDate Inc. Dr Kang reported receiving grants from PTC Therapeutics. Dr Mah reported receiving grants from ReveraGen BioPharma Inc, Pfizer, Italfarmaco, Sarepta Therapeutics, Catabasis Pharmaceuticals, NS Pharma, and PTC Therapeutics. Dr Mongini reported receiving personal fees from Sanofi Genzyme, Biogen Inc, Sarepta Therapeutics, Spark, Roche, and Novartis. Dr Ricci reported receiving grants from the University of Rochester. Dr von der Hagen reported receiving personal fees from PTC Therapeutics Germany GmbH and Pfizer Pharma GmbH and receiving grants from Sarepta Therapeutics. Dr Finkel reported receiving grants from ReveraGen BioPharma Inc, the Muscular Dystrophy Association, the National Institutes of Health, Capricor Therapeutics, Catabasis Pharmaceuticals, Eli Lilly, PTC Therapeutics, Santhera Pharmaceuticals, Sarepta Therapeutics, and Summit Therapeutics. Ms O’Reardon reported receiving grants from Nemours Children’s Hospital. Dr Wicklund reported receiving personal fees from Affinia Therapeutics, Amicus Therapeutics, Edgewise Therapeutics, ML Bio Solutions, Sarepta Therapeutics, and Sanofi-Genzyme. Dr Kumar reported receiving grants from PTC Therapeutics, Sarepta Therapeutics, AveXis (now Novartis Gene Therapies), the Muscular Dystrophy Association, and FibroGen Inc and receiving personal fees from Sarepta Therapeutics, Audentes Therapeutics, AveXis, Roche, Genetech, PTC Therapeutics, Biogen Inc, and the American Academy of Pediatrics. Dr McDonald reported receiving grants from Capricor Therapeutics, Catabasis Pharmaceuticals, FibroGen Inc, Pfizer, PTC Therapeutics, ReveraGen BioPharma Inc, Santhera Pharmaceuticals, the University of Rochester, and Sarepta Therapeutics and receiving personal fees from PTC Therapeutics, Sarepta Therapeutics, Astellas, Avidity Biosciences, BioMarin Pharmaceutical Inc, Capricor Therapeutics, Catabasis Pharmaceuticals, Edgewise Therapeutics, Eli Lilly, Entrada Therapeutics, Epirium Bio, FibroGen Inc, Hoffmann-La Roche, Italfarmaco, Marathon Pharmaceuticals, Pfizer, PTC Therapeutics, Sarepta Therapeutics, and Santhera Pharmaceuticals. Dr Henricson reported receiving personal fees from PTC Therapeutics and Santhera Pharmaceuticals. Dr Schara-Schmidt reported receiving personal fees from Sarepta Therapeutics, Santhera Pharmaceuticals, Roche, PTC Therapeutics, and Pfizer. Dr Wilichowski reported receiving grants from the University of Rochester. Dr Statland reported receiving grants from the National Institutes of Health, the FSHD Society, the FSHD Canada Foundation, and the Friends of FSH Research and receiving personal fees from Fulcrum Therapeutics, Dyne Therapeutics Inc, Avidity Biosciences, Mitsubishi Tanabe Pharma, ML Bio Solutions, and Amylyx Pharmaceuticals. Dr Campbell reported receiving personal fees from Acceleron Pharma, AMO Pharma, BioMarin Pharmaceutical Inc, Bristol Myers Squibb, Eli Lily, Biogen Inc, Pfizer, Roche, PTC Therapeutics, Sarepta Therapeutics, Cytokinetics, Wave Therapeutics, Catabasis Pharmaceuticals, Edgewise Therapeutics, and Solid Biosciences and receiving grants from PTC Therapeutics, Genzyme, and Biogen Inc. Dr G. Vita reported receiving grants from Sarepta Therapeutics, Santhera Pharmaceuticals, Italfarmaco, and Wave Therapeutics. Dr G. L. Vita reported receiving grants from Sarepta Therapeutics, Santhera Pharmaceuticals, Italfarmaco, and Wave Therapeutics. Dr Howard reported receiving grants from Alexion Pharmaceuticals, Ra Pharmaceuticals (now UCB), Argenx BVBA, and Millennium Pharmaceuticals Inc and receiving personal fees from Alexion Pharmaceuticals, Argenx BVBA, Ra Pharmaceuticals, Immunovant Inc, Regeneron Pharmaceuticals, Toleranzia, and Viela Bio Inc (now Horizon Therapeutics). Dr Hughes reported receiving personal fees from PTC Therapeutics, Novartis, Santhera Pharmaceuticals, Summit Therapeutics, and Roche. Dr Pegoraro reported receiving personal fees from PTC Therapeutics, Sarepta Therapeutics, Epirium Bio, Roche, Biogen Inc, UCB, and Alexion Pharmaceuticals and receiving grants and nonfinancial support from Santhera Pharmaceuticals. Dr Bello reported serving on boards for Edgewise Therapeutics, PTC Therapeutics, and Sarepta Therapeutics; receiving grant support from Santhera Pharmaceuticals; and receiving personal fees from PTC Therapeutics and Epirium Bio. Dr Burnette reported serving on working groups or boards for PTC Therapeutics, Sarepta Therapeutics, and SteroTherapeutics LLC. Dr Thangarajh reported serving as a consultant to Sarepta Therapeutics. Dr Griggs reported serving as a consultant to Strongbridge and Stealth Pharmaceuticals; receiving personal fees from Solid Biosciences and Elsevier; serving as chair of the research advisory committee and is a board member of the American Brain Foundation; and serving on the executive committee of the Muscle Study Group, which receives support for its activities from pharmaceutical companies. No other disclosures were reported.
http://www.ncbi.nlm.nih.gov/pubmed/19488064
1. Pharmacogenomics J. 2009 Dec;9(6):411-8. doi: 10.1038/tpj.2009.22. Epub 2009 Jun 2. Corticosteroid effects on blood gene expression in Duchenne muscular dystrophy. Lit L(1), Sharp FR, Apperson M, Liu DZ, Walker WL, Liao I, Xu H, Ander BP, Wong B. Author information: (1)Department of Neurology, MIND Institute, University of California at Davis, Sacramento, CA 95817, USA. [email protected] Though Deflazacort and prednisone improve clinical endpoints in Duchenne muscular dystrophy (DMD) patients, Deflazacort produces fewer side effects. As mechanisms of improvement and side effect differences remain unknown, we evaluated effects of corticosteroid administration on gene expression in blood of DMD patients. Whole blood was obtained from 14 children and adolescents with DMD treated with corticosteroids (DMD-STEROID) and 20 DMD children and adolescents naïve to corticosteroids (DMD). The DMD-STEROID group was further subdivided into Deflazacort and prednisone groups. Affymetrix U133 Plus 2.0 expression microarrays were used to evaluate mRNA expression. Expression of 524 probes changed with corticosteroids, including genes in iron trafficking and the chondroitin sulfate biosynthesis pathway. Deflazacort compared with prednisone yielded 508 regulated probes, including many involved in adipose metabolism. These genes and pathways help explain mechanisms of efficacy and side effects of corticosteroids, and could provide new treatment targets for DMD and other neuromuscular disorders. DOI: 10.1038/tpj.2009.22 PMID: 19488064 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35928819
1. Front Immunol. 2022 Jul 19;13:909008. doi: 10.3389/fimmu.2022.909008. eCollection 2022. Role of Bispecific Antibodies in Relapsed/Refractory Diffuse Large B-Cell Lymphoma in the CART Era. González Barca E(1). Author information: (1)Hematology Department, Catalan Institute of Oncology, University of Barcelona, IDIBELL, Barcelona, Spain. Diffuse large B-cell lymphoma is an aggressive and biologically heterogeneous disease. R-CHOP is the standard first line therapy and cures more than 60% of patients. Salvage high-dose chemotherapy with autologous stem cell transplant remains the standard second-line treatment for relapsed or refractory patients, and recently, three CD19 chimeric antigen receptor T cells (CART) cell products have been approved beyond 2 prior lines of systemic therapy. Nevertheless, some patients are not eligible for transplant or CARTs, or progress after these treatments. In this context, IgG-like bispecific antibodies (BsAbs) have been designed to treat B-cell lymphomas. They combine two different monospecific antigen-binding regions that target CD20 on B cells and engage T cells via CD3 in a 1:1 or 2:1 CD20:CD3 antigen binding fragment (Fab) format. The results of different phase 1 trials with BsAbs, including mosunetuzumab, glofitamab, epcoritamab and odeonextamab, have been recently published. They are infused intravenously or subcutaneously, and have a favorable toxicity profile, with reduced cytokine release syndrome and neurological toxicity. Moreover, these BsAbs have demonstrated very promising efficacy in B-cell lymphomas, including in aggressive lymphomas. New trials are currently ongoing to confirm BsAbs efficacy and tolerability, as well as to explore its efficacy in different lines of therapy or in combination with other drugs. Copyright © 2022 González Barca. DOI: 10.3389/fimmu.2022.909008 PMCID: PMC9344863 PMID: 35928819 [Indexed for MEDLINE] Conflict of interest statement: EG declares having received lecture fees and advisory board fees from Janssen, Abbvie, Gilead, Kiowa, EUSAPharma, Incyte, Lilly, Beigene, Novartis, Abbvie, Takeda, and Roche.
http://www.ncbi.nlm.nih.gov/pubmed/35803286
1. Lancet Oncol. 2022 Aug;23(8):1055-1065. doi: 10.1016/S1470-2045(22)00335-7. Epub 2022 Jul 5. Safety and efficacy of mosunetuzumab, a bispecific antibody, in patients with relapsed or refractory follicular lymphoma: a single-arm, multicentre, phase 2 study. Budde LE(1), Sehn LH(2), Matasar M(3), Schuster SJ(4), Assouline S(5), Giri P(6), Kuruvilla J(7), Canales M(8), Dietrich S(9), Fay K(10), Ku M(11), Nastoupil L(12), Cheah CY(13), Wei MC(14), Yin S(14), Li CC(14), Huang H(15), Kwan A(14), Penuel E(14), Bartlett NL(16). Author information: (1)Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA, USA. (2)Department of Medical Oncology, BC Cancer Centre for Lymphoid Cancer, Vancouver, BC, Canada; Department of Medicine, The University of British Columbia, Vancouver, BC, Canada. (3)Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA. (4)Lymphoma Translational Research, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA. (5)Division of Hematology, Jewish General Hospital, Montreal, QC, Canada. (6)Department of Haematology, Royal Adelaide Hospital, Adelaide, SA, Australia. (7)Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada. (8)Servicio de Hematología, Hospital Universitario La Paz, Madrid, Spain. (9)Department of Internal Medicine V: Hematology, Oncology and Rheumatology, Heidelberg University, Heidelberg, Germany. (10)Department of Haematology, Royal North Shore Hospital, Sydney, NSW, Australia; Department of Haematology, St Vincent's Hospital, University of Melbourne, Melbourne, VIC, Australia. (11)Department of Haematology, St Vincent's Hospital, University of Melbourne, Melbourne, VIC, Australia. (12)Department of Lymphoma/Myeloma, MD Anderson Cancer Center, Houston, TX, USA. (13)Linear Clinical Research, Sir Charles Gairdner Hospital, Perth, WA, Australia; Lymphoma Translational Research, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA; Department of Haematology, The University of Western Australia, Perth, WA, Australia. (14)Genentech, San Francisco, CA, USA. (15)F Hoffmann-La Roche, Mississauga, ON, Canada. (16)Division of Oncology, Siteman Cancer Center, Washington University School of Medicine, St Louis, MO, USA. Electronic address: [email protected]. Comment in Lancet Oncol. 2022 Aug;23(8):967-969. doi: 10.1016/S1470-2045(22)00385-0. BACKGROUND: Mosunetuzumab is a CD20 × CD3 T-cell-engaging bispecific monoclonal antibody that redirects T cells to eliminate malignant B cells. In a phase 1 study, mosunetuzumab was well tolerated and active in patients with relapsed or refractory B-cell lymphoma. We, therefore, aimed to evaluate the safety and anti-tumour activity of fixed-duration mosunetuzumab in patients with relapsed or refractory follicular lymphoma who had received two or more previous therapies. METHODS: We conducted a single-arm, multicentre, phase 2 study at 49 centres in seven countries (Australia, Canada, Germany, South Korea, Spain, UK, and USA). All patients were aged 18 years or older with histologically confirmed follicular lymphoma (grade 1-3a) and an Eastern Cooperative Oncology Group performance status of 0-1. Patients had disease that was relapsed or refractory to two or more previous lines of treatment, including an anti-CD20 therapy and an alkylating agent. Intravenous mosunetuzumab was administered in 21-day cycles with cycle 1 step-up dosing: 1 mg on cycle 1 day 1, 2 mg on cycle 1 day 8, 60 mg on cycle 1 day 15 and cycle 2 day 1, and 30 mg on day 1 of cycle 3 and onwards. Patients with a complete response by investigator assessment using the International Harmonisation Project criteria completed treatment after cycle 8, whereas patients with a partial response or stable disease continued treatment for up to 17 cycles. The primary endpoint was independent review committee-assessed complete response rate (as best response) in all enrolled patients; the primary efficacy analysis compared the observed IRC-assessed complete response rate with a 14% historical control complete response rate in a similar patient population receiving the pan class I PI3K inhibitor copanlisib. Safety was assessed in all enrolled patients. This study is registered with ClinicalTrials.gov, number NCT02500407, and is ongoing. FINDINGS: Between May 2, 2019, and Sept 25, 2020, we enrolled 90 patients. As of the data cutoff date (Aug 27, 2021), the median follow-up was 18·3 months (IQR 13·8-23·3). According to independent review committee assessment, a complete response was recorded in 54 patients (60·0% [95% CI 49·1-70·2]). The observed complete response rate was significantly higher than the historical control complete response rate with copanlisib of 14% (p<0·0001), thereby meeting the primary study endpoint. Cytokine release syndrome was the most common adverse event (40 [44%] of 90 patients) and was predominantly grade 1 (23 [26%] of 90) and grade 2 (15 [17%]), and primarily confined to cycle 1. The most common grade 3-4 adverse events were neutropenia or neutrophil count decreased (24 [27%] of 90 patients), hypophosphataemia (15 [17%]), hyperglycaemia (seven [8%]), and anaemia (seven [8%]). Serious adverse events occurred in 42 (47%) of 90 patients. No treatment-related grade 5 (ie, fatal) adverse event occurred. INTERPRETATION: Fixed-duration mosunetuzumab has a favourable safety profile and induces high rates of complete remissions, allowing potential administration as an outpatient regimen, in patients with relapsed or refractory follicular lymphoma and two or more previous therapies. FUNDING: F Hoffmann-La Roche and Genentech. Copyright © 2022 Elsevier Ltd. All rights reserved. DOI: 10.1016/S1470-2045(22)00335-7 PMID: 35803286 [Indexed for MEDLINE] Conflict of interest statement: Declaration of interests LEB reports consulting fees from Genentech, ADC Therapeutics, Merck, AstraZeneca, and Amgen; and participation on a data safety monitoring board for Ziopharm Oncology. LHS reports research grants from Roche/Genentech and Teva; consulting fees from AbbVie, Acerta, Amgen, AstraZeneca, Celgene/Bristol-Myers Squibb (BMS), Gilead/Kite/Incyte, Janssen, Roche/Genentech, MorphoSys, Sandoz, and TG therapeutics; and honoraria from AbbVie, Acerta, Amgen, AstraZeneca, Celgene/BMS, Gilead/Kite/Incyte, Janssen, Roche/Genentech, MorphoSys, Sandoz, and TG therapeutics. MM reports research grants from AstraZeneca, Genentech, Janssen, Roche, Bayer, IGM Biosciences, Pharmacyclics, and Seattle Genetics; payment or honoraria from lectures, presentations, speaker's bureaus or manuscript writing, or educational events from ADC Therapeutics, Bayer, Daiichi Sankyo, MEI Pharma, Genentech, Seattle Genetics, Epizyme, IMV Therapeutics, Janssen, Pharmacyclics, and Roche; payment from expert testimony from Bayer; and owns stock or stock options in Merck. SJS reports grants or contracts from Genentech, AbbVie, Acerta, AstraZeneca, Celgene/BMS/Juno, Incyte, Merck, Novartis, TG Therapeutics, and Theredex; consulting fees from AstraZeneca, BeiGene, Celgene/BMS/Juno, Genentech/Roche, Genmab, Incyte, Janssen, MorphoSys, Mustang Biotech, Novartis, and Regeneron; payment or honoraria from lectures, presentations, speaker's bureaus or manuscript writing, or educational events for Incyte, Novartis, and Takeda; participation on a monitoring board or advisory board for AstraZeneca, BeiGene, Celgene/BMS/Juno, Genentech/Roche, Genmab, Incyte, Janssen, MorphoSys, Mustang Biotech, Novartis, and Regeneron; a role in board, society, committee, or advocacy groups for Genentech, Legend Biotech, Novartis, and Nordic Nanovector; and research support for Genentech and Merck. SA reports research grants from AbbVie, Roche, Genentech, Takeda, Lilly, and Merck; speaker's bureau from Pfizer; and is chair of the haematology group (unpaid) of the Canadian Cancer Trials Group. JK reports research grants from Roche, AstraZeneca, and Merck; consulting fees from AbbVie, Antengene, BMS, Gilead, Karyopharm, Medison Ventures, Merck, Roche, and Seattle Genetics; payment or honoraria from lectures, presentations, and educational events for AbbVie, Amgen, AstraZeneca, BMS, Gilead, Incyte, Janssen, Karyopharm, Merck, Novartis, Pfizer, Roche, and Seattle Genetics; participation in a data safety monitoring board for Karyopharm; and participation as chair, scientific advisory board, and director in the board of directors for Lymphoma Canada. MC reports consulting fees from BeiGene, BMS, Incyte, Janssen, Kite, Kyowa, Miltenyi Biotec, Novartis, Roche, and Takeda; payment from lectures, presentations, speaker's bureaus or manuscript writing, or educational events from Amgen, Eusa Pharma, Janssen, Kite, Kyowa, Roche, and Takeda; and meeting attendance or travel support, or both, from Kite, Janssen, Roche, Sanofi, and Takeda. LN reports grants from BMS, Caribou Biosciences, Epizyme, Gilead/Kite, Janssen, IGM Biosciences, Takeda, and TG Therapeutics; payment or honoraria from lectures, presentations, speaker's bureaus or manuscript writing, or educational events from Genentech, Gilead/Kite, and Takeda; meeting attendance or travel, or both, support from Genentech; and participation in data safety monitoring board or advisory board for ADC Therapeutics, Bayer, Epizyme, BMS, MorphoSys, Novartis, Genentech, Takeda, MEI, DeNovo, and TG Therapeutics. CYC reports payment or honoraria from lectures, presentations, speaker's bureaus or manuscript writing, or educational events for Janssen, AstraZeneca, Roche, and Beigene; and participation in data safety monitoring board or advisory board for Roche, Janssen, TG Therapeutics, AstraZeneca, Lilly, and Gilead. MCW reports meeting attendance or travel support, or both, from Roche/Genentech; and stocks and stock options in Roche. SY reports meeting attendance or travel support, or both, for Genentech; patents planned, issued, or pending from Genentech; and stocks and stock options in Genentech. C-CL reports stocks and stock options in Roche; and patents planned, issued, or pending for Genentech. AK reports stocks or stock options in Roche. EP reports stocks or stock options in Genentech. NLB reports research funding from ADC Therapeutics, Affimed, Autolus, BMS, Celgene, Forty Seven, Immune Design, Janssen, Kite Pharma, Merck, Millennium, Pfizer, Pharmacyclics, Roche/Genentech, and Seattle Genetics. All other authors declare no competing interests.
http://www.ncbi.nlm.nih.gov/pubmed/35947358
1. Drugs. 2022 Jul;82(11):1229-1234. doi: 10.1007/s40265-022-01749-5. Mosunetuzumab: First Approval. Kang C(1). Author information: (1)Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand. [email protected]. Mosunetuzumab (Lunsumio®), an anti-CD20/CD3 T-cell engaging bispecific antibody, is being developed by Roche for the treatment of relapsed or refractory follicular lymphoma. Mosunetuzumab was recently conditionally approved in the EU for the treatment of relapsed or refractory follicular lymphoma in adults who have received at least two prior systemic therapies. This article summarizes the milestones in the development of mosunetuzumab leading to this first approval for relapsed or refractory follicular lymphoma. © 2022. The Author(s), under exclusive licence to Springer Nature Switzerland AG. DOI: 10.1007/s40265-022-01749-5 PMID: 35947358 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/35182296
1. Curr Treat Options Oncol. 2022 Feb;23(2):155-170. doi: 10.1007/s11864-021-00925-1. Epub 2022 Feb 19. Bispecific Antibodies for Non-Hodgkin Lymphoma Treatment. Bock AM(1), Nowakowski GS(1), Wang Y(2). Author information: (1)Division of Hematology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA. (2)Division of Hematology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA. [email protected]. While there have been numerous advances in the field of non-Hodgkin lymphoma (NHL) over the last decade, relapsed and/or refractory (R/R) NHL remains a challenge and an area with unmet needs. T-cell redirecting immunotherapeutic approaches including chimeric antigen receptor (CAR) T-cells and bispecific antibodies (BsAbs) have the potential to revolutionize NHL therapy. BsAbs target CD3 on T-cells and CD19 or CD20 on malignant B-cells and have shown promises as a novel immunotherapy for NHL. The development of CD19 × CD3 BsAbs such as blinatumomab was met with significant challenges due to dose-limiting neurologic side effects. However, several CD20 × CD3 BsAbs including odronextamab, mosunetuzumab, glofitamab, and epcoritamab emerged recently. They have favorable toxicity profiles, with reduced cytokine release syndrome and neurotoxicity. In addition, all these BsAbs have demonstrated very promising efficacy in R/R NHL. With expansion and registrational studies actively ongoing, approvals of these agents for R/R NHL are anticipated in the near future. Some important questions pertinent to future clinical development of BsAbs include when and how to best utilize BsAbs in the management of R/R NHL, whether there is a role of BsAbs in treatment-naïve NHL, and how to combine BsAbs with other therapies. For example, whether BsAbs can be combined with cytotoxic chemotherapy effectively remains to be seen. A plethora of clinical studies will be needed to help address these questions, some of which are already ongoing. In addition, how do BsAbs compare to CAR T-cell therapy and how to choose and sequence between BsAbs and CAR T-cell therapy need to be addressed. While many of these critical questions remain to be answered in clinical studies, we believe the future of BsAbs in the NHL is very bright. © 2022. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature. DOI: 10.1007/s11864-021-00925-1 PMID: 35182296 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/20678798
1. Mol Immunol. 2010 Nov-Dec;48(1-3):14-25. doi: 10.1016/j.molimm.2010.07.009. Epub 2010 Aug 1. Mast cells: emerging sentinel innate immune cells with diverse role in immunity. Kumar V(1), Sharma A. Author information: (1)Department of Microbiology, Panjab University, Chandigarh-160014, India. vij [email protected] Mast cells are phylogenetically old innate immune cells with less recognition in normal function of immune system as no such disease has been observed in humans due to their deficiency or inadequate function. Earlier mast cells were only known for their important role in the type 1 allergic reactions (i.e. anaphylaxis or some contact hypersensitivity reactions) due to release of various biochemical mediators (i.e. cytokines, chemokines, lipid mediators, proteases and biologic amines). Several studies indicated that they do not only come in action upon binding of IgE to its corresponding receptors expressed by them but also play an important role in host immunity. Recent development in understanding the mast cell biology has established various important roles of these cells in regulating both innate as well as adaptive immune response under normal or pathophysiological conditions (i.e. acute or chronic bacterial or parasitic infections, various autoimmune disease, pregnancy, etc.). Present review is designed to accommodate up to date information regarding mast cell development (i.e. factors governing mast cell development and their homing to various compartments (i.e. skin, lungs, intestine, uterus, etc.) along with their role in innate immunity, human pregnancy and future immunomodulatory approach comprising of targeting mast cells. Copyright © 2010 Elsevier Ltd. All rights reserved. DOI: 10.1016/j.molimm.2010.07.009 PMID: 20678798 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/23855441
1. J Vet Emerg Crit Care (San Antonio). 2013 Jul-Aug;23(4):377-94. doi: 10.1111/vec.12066. Epub 2013 Jul 15. Anaphylaxis in dogs and cats. Shmuel DL(1), Cortes Y. Author information: (1)Oradell Animal Hospital, Emergency and Critical Care, 580 Winters Avenue, Paramus, NJ 07652, USA. [email protected] Comment in J Vet Emerg Crit Care (San Antonio). 2014 Mar-Apr;24(2):134; discussion 134. doi: 10.1111/vec.12149. OBJECTIVE: To review and summarize current information regarding the pathophysiology and clinical manifestations associated with anaphylaxis in dogs and cats. The etiology, diagnosis, treatment, and prognosis is discussed. ETIOLOGY: Anaphylaxis is a systemic, type I hypersensitivity reaction that often has fatal consequences. Many of the principal clinical manifestations involve organs where mast cell concentrations are highest: the skin, the lungs, and the gastrointestinal tract. Histamine and other deleterious inflammatory mediators promote vascular permeability and smooth muscle contraction; they are readily released from sensitized mast cells and basophils challenged with antigen. Anaphylaxis may be triggered by a variety of antigens including insect and reptile venom, a variety of drugs, vaccines, and food. DIAGNOSIS: Anaphylaxis is a clinical diagnosis made from a collection of signs and symptoms. It is most commonly based on pattern recognition. Differential diagnoses include severe asthma, pheocromocytoma, and mastocytosis. THERAPY: Epinephrine is considered the drug of choice for the treatment of anaphylaxis. It acts primarily as a vasopressor in improving hemodynamic recovery. Adjunctive treatments include fluid therapy, H1 and H2 antihistamines, corticosteroids, and bronchodilators; however, these do not substitute for epinephrine. PROGNOSIS: Prognosis depends on the severity of the clinical signs. The clinical signs will vary among species and route of exposure. The most severe clinical reactions are associated when the antigen is administered parenterally. © Veterinary Emergency and Critical Care Society 2013. DOI: 10.1111/vec.12066 PMID: 23855441 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/17128671
1. Lijec Vjesn. 2006 Sep-Oct;128(9-10):317-21. [Anaphylactic reaction as a side-effect of administration of general anesthetics and neuromuscular blocking agents]. [Article in Croatian] Milavec D(1), Husedzinović I. Author information: (1)Klinika za anesteziologiju, reanimatologiju i intenzivno lijecenje, Klinicka bolnica Dubrava, Zagreb. The incidence of anaphylactic reactions during anesthesia is between 1:5000 and 1:25000 anesthetics. During the IgE-mediated anaphylactic reaction mast cells release proteases such as tryptase, histamine and vasoactive mediators. The release of mediators from the mast cells and basophils is responsible for the immediate clinical manifestations of anaphylaxis. Anaphylactoid reactions can be caused directly by a release of histamine and other mediators from mast cells and basophils and they don't depend on interaction of IgE antibodies with antigen. The most frequent agents that cause anaphylactic and anaphylactoid reactions during anesthesia are neuromuscular blocking agents (among them the highest percent refers to rocuronium and succinylcholine), some general anesthetics, antibiotics, blood and blood products, opioids and latex. Increased tryptase concentration in serum is a marker for systemic mast cell activation. Skin tests (in vivo) are used for verification of specific hypersensitivity to drugs in patients after anaphylactic reaction. In vitro tests prove the presence of specific IgE antibodies for drugs. The plan for the treatment of anaphylactic reactions must be established before the event. Airway maintenance, 100% oxygen administration, intravascular volume expansion and epinephrine are essential to treat the hypotension and hypoxia that result from vasodilatation, increased capillary permeability and bronchospasm. As soon as the diagnosis has been made the adrenalin should be given intravenously 1 to 3 ml of 1:10000 aqueous solution (0.1 mg/ml) over 10 minutes. Prevention is possible with methylprednisolone 125 mg i.v. 1 hour before administering of anesthetics and neuromuscular blocking agents with or without antihistaminic chlorpiramine-chloride 1 amp i.v. few minutes before anesthesia. PMID: 17128671 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/19082920
1. Arch Immunol Ther Exp (Warsz). 2008 Nov-Dec;56(6):385-99. doi: 10.1007/s00005-008-0041-2. Epub 2008 Dec 1. Positive and negative regulatory mechanisms in high-affinity IgE receptor-mediated mast cell activation. Roth K(1), Chen WM, Lin TJ. Author information: (1)Department of Microbiology and Immunology, Dalhousie University, 5850 University Ave., Halifax, NS, Canada B3K6R8. Mast cells are important effector cells in allergic inflammatory reactions. The aggregation of the high-affinity IgE receptor (FcepsilonRI) on the surface of mast cells initiates a complex cascade of signaling events that ultimately leads to the release of various mediators involved in allergic inflammation and anaphylactic reactions. The release of these mediators is tightly controlled by signaling pathways that are propagated through the cell by specific phosphorylation and dephosphorylation events. These events are controlled by protein kinases and protein phosphatases which either positively or negatively regulate the propagation of the signal through the cell. This review summarizes the role of both positive and negative regulators of FcepsilonRI-induced mast cell activation. DOI: 10.1007/s00005-008-0041-2 PMID: 19082920 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/32276688
1. J Allergy Clin Immunol Pract. 2020 Apr;8(4):1196-1201. doi: 10.1016/j.jaip.2019.10.048. Idiopathic Anaphylaxis: A Form of Mast Cell Activation Syndrome. Giannetti MP(1), Akin C(2), Castells M(3). Author information: (1)Brigham and Women's Hospital, Division of Allergy and Clinical Immunology, Boston, Mass; Harvard Medical School, Boston, Mass. Electronic address: [email protected]. (2)Division of Allergy and Immunology, University of Michigan, Ann Arbor, Mich. (3)Brigham and Women's Hospital, Division of Allergy and Clinical Immunology, Boston, Mass; Harvard Medical School, Boston, Mass. Idiopathic anaphylaxis is a condition caused by paroxysmal episodes of sudden-onset multiorgan involvement variably including laryngeal edema, urticaria, bronchoconstriction, dyspnea, hypoxia, abdominal pain, nausea, vomiting, diarrhea, and hypotension. Rarely, the episodes can lead to cardiovascular collapse and death in the absence of a clear trigger, especially in the presence of other cardiovascular comorbidities. Elevated mast cell mediators such as tryptase and histamine have been reported during episodes, and mast cells are considered the primary cells responsible for driving anaphylaxis in humans. Basophils also secrete histamine and LTC4 when activated and theoretically can contribute to symptoms. As our understanding of mast cell disorders continue to grow, the classification for these disorders evolves. The purpose of this article was 2-fold: to review the epidemiology, clinical manifestations, and diagnosis of idiopathic anaphylaxis and to discuss the classification of idiopathic anaphylaxis within the broader context of mast cell activation disorders. Copyright © 2019 American Academy of Allergy, Asthma & Immunology. Published by Elsevier Inc. All rights reserved. DOI: 10.1016/j.jaip.2019.10.048 PMID: 32276688 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/25841551
1. Immunol Allergy Clin North Am. 2015 May;35(2):277-85. doi: 10.1016/j.iac.2015.01.010. Mast cell activation syndromes presenting as anaphylaxis. Akin C(1). Author information: (1)Department of Medicine, Division of Rheumatology, Immunology and Allergy, Mastocytosis Center, Brigham and Women's Hospital, Harvard Medical School, One Jimmy Fund Way, Room 616D Boston, MA 02115, USA. Electronic address: [email protected]. Anaphylaxis results from severe systemic mast cell activation. In addition to IgE-mediated and physical triggers, it may occur with a clonal mast cell disease and in an idiopathic fashion without clear provoking factors. Disorders of mast cell activation are classified into primary (clonal), secondary, and idiopathic. Mast cell activation syndrome (MCAS) is a multisystem disorder characterized by objective documentation of elevated mast cell mediators during attacks and a favorable response to antimediator therapy. It should be considered in the differential diagnosis of patients presenting with recurrent anaphylaxis without a clear cause. This article discusses the diagnosis of MCAS. Copyright © 2015 Elsevier Inc. All rights reserved. DOI: 10.1016/j.iac.2015.01.010 PMID: 25841551 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/28798744
1. Front Immunol. 2017 Jul 26;8:846. doi: 10.3389/fimmu.2017.00846. eCollection 2017. The Mast Cell, Contact, and Coagulation System Connection in Anaphylaxis. Guilarte M(1)(2), Sala-Cunill A(1)(2), Luengo O(1)(2), Labrador-Horrillo M(1)(2), Cardona V(1)(2). Author information: (1)Allergy Section, Internal Medicine Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain. (2)VHIR Institut de Recerca Vall d'Hebron, Barcelona, Spain. Anaphylaxis is the most severe form of allergic reaction, resulting from the effect of mediators and chemotactic substances released by activated cells. Mast cells and basophils are considered key players in IgE-mediated human anaphylaxis. Beyond IgE-mediated activation of mast cells/basophils, further mechanisms are involved in the occurrence of anaphylaxis. New insights into the potential relevance of pathways other than mast cell and basophil degranulation have been unraveled, such as the activation of the contact and the coagulation systems. Mast cell heparin released upon activation provides negatively charged surfaces for factor XII (FXII) binding and auto-activation. Activated FXII, the initiating serine protease in both the contact and the intrinsic coagulation system, activates factor XI and prekallikrein, respectively. FXII-mediated bradykinin (BK) formation has been proven in the human plasma of anaphylactic patients as well as in experimental models of anaphylaxis. Moreover, the severity of anaphylaxis is correlated with the increase in plasma heparin, BK formation and the intensity of contact system activation. FXII also activates plasminogen in the fibrinolysis system. Mast cell tryptase has been shown to participate in fibrinolysis through plasmin activation and by facilitating the degradation of fibrinogen. Some usual clinical manifestations in anaphylaxis, such as angioedema or hypotension, or other less common, such as metrorrhagia, may be explained by the direct effect of the activation of the coagulation and contact system driven by mast cell mediators. DOI: 10.3389/fimmu.2017.00846 PMCID: PMC5526842 PMID: 28798744
http://www.ncbi.nlm.nih.gov/pubmed/26857018
1. Curr Allergy Asthma Rep. 2016 Mar;16(3):20. doi: 10.1007/s11882-016-0598-5. Mast Cells and Anaphylaxis. Lieberman P(1)(2), Garvey LH(3). Author information: (1)Divisions of Allergy and Immunology, Departments of Medicine and Pediatrics, University of Tennessee, Memphis, TN, USA. [email protected]. (2), 7205 Wolf River Blvd., Suite 200, Germantown, TN, 38138-1777, USA. [email protected]. (3)Allergy Clinic UA-816, Department of Dermato-allergology, Copenhagen University Hospital, Gentofte, Kildegårdsvej 28, DK-2900, Hellerup, Denmark. [email protected]. For half a century, it has been known that the mast cell is the cell responsible for the majority of anaphylactic events. Its mediators, taken as a whole, are capable of producing all of the clinical manifestations of these events. With the discovery of immunoglobulin E (IgE), it was originally felt that the vast majority of anaphylactic episodes were due to antigen coupling with two cell-bound IgE molecules. More recently it has been learned that many episodes are produced by direct activation of mast cells, not involving antigen binding to IgE, and that monomeric IgE under certain conditions can also cause degranulation. Of note--in regard to antigen independent degranulation--are recent reports that the human G-protein-coupled receptor, MRGPRX2, may be the receptor for many drugs and cationic proteins capable of producing direct mast cell degranulation and anaphylactic events. DOI: 10.1007/s11882-016-0598-5 PMID: 26857018 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/33832694
1. J Allergy Clin Immunol. 2021 Apr;147(4):1123-1131. doi: 10.1016/j.jaci.2021.02.012. Non-IgE-mediated anaphylaxis. Cianferoni A(1). Author information: (1)Perelman School of Medicine, University of Pennsylvania, Allergy and Immunology Division, The Children's Hospital of Philadelphia, Philadelphia, Pa. Electronic address: [email protected]. Anaphylaxis is a rapidly evolving, acute, life-threatening reaction that occurs rapidly on contact with a trigger. Anaphylaxis is classically defined as an allergen-driven process that induces specific IgE and the activation of mast cells and basophils through the cross-linking of IgE receptors. However, it is clear that non-IgE-mediated pathways can induce symptoms indistinguishable from those of classic anaphylaxis, and their activation could explain the severity of IgE-mediated anaphylaxis. Indeed, mast cells and basophils can be activated by antibodies against IgE or their receptors, by molecules such as anaphylatoxins, or through G-coupled receptors. Some other allergens can induce antibodies of class IgG that can activate neutrophils to produce a molecule similar to histamine to induce anaphylaxis. Finally, some inflammatory mediators such as bradykinin or prostaglandin can also modulate mast cell and basophil activation as well as directly cause vasodilation and bronchoconstriction, resulting in anaphylaxis-like reactions. Copyright © 2021 American Academy of Allergy, Asthma & Immunology. Published by Elsevier Inc. All rights reserved. DOI: 10.1016/j.jaci.2021.02.012 PMID: 33832694 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/20519881
1. Chem Immunol Allergy. 2010;95:45-66. doi: 10.1159/000315937. Epub 2010 Jun 1. Anaphylaxis: mechanisms of mast cell activation. Kalesnikoff J, Galli SJ. Anaphylaxis is a severe systemic allergic response that is rapid in onset and potentially lethal, and that typically is induced by an otherwise innocuous substance. In IgE-dependent and other examples of anaphylaxis, tissue mast cells and circulating basophilic granulocytes (basophils) are thought to represent major (if not the major) sources of the biologically active mediators that contribute to the pathology and, in unfortunate individuals, fatal outcome, of anaphylaxis. In this chapter, we will describe the mechanisms of mast cell (and basophil) activation in anaphylaxis, with a focus on IgE-dependent activation, which is thought to be responsible for most examples of antigen-induced anaphylaxis in humans. We will also discuss the use of mouse models to investigate the mechanisms that can contribute to anaphylaxis in that species in vivo, and the relevance of such mouse studies to human anaphylaxis. Copyright 2010 S. Karger AG, Basel. DOI: 10.1159/000315937 PMID: 20519881 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31690393
1. Allergy Asthma Proc. 2019 Nov 1;40(6):453-456. doi: 10.2500/aap.2019.40.4270. Anaphylaxis. Watts MM, Marie Ditto A. Anaphylaxis is a sudden onset, immediate reaction that implies a risk of death. Think of a "rule of 2s" for anaphylaxis, which implies that reactions usually begin within 2 minutes to 2 hours after injection, infusion, ingestion, contact, or inhalation. Fatalities can be from asphyxiation from laryngeal or oropharyngeal swelling, collapse from hypotensive shock, cardiac arrest, or acute severe bronchoconstriction that causes respiratory failure and arrest. When there is activation of mast cells and basophils in anaphylaxis, chemical mediators are detectable. The preformed mediators from mast cells include histamine, tryptase, carboxypeptidase A, and proteoglycans (heparin, chondroitin sulfates). Newly synthesized mediators include prostaglandin D₂, leukotriene D₄, and platelet activating factor. Crucial actions of the mediators include an abrupt increase in vascular permeability, vascular smooth muscle relaxation, and bronchial smooth muscle contraction. Anaphylaxis can be classified into immunologic, nonimmunologic, or idiopathic based on the associated mechanism. For example, immunologic causes of anaphylaxis are those mediated by immunoglobulin E (IgE) antibodies acting through the FcεR I (foods, insect venom, 32 β-lactam antibiotics), whereas non-IgE immunologic anaphylaxis is mediated without the presence of anti-allergen IgE antibodies or via FcεRI activation (radiographic contrast material). Nonimmunologic anaphylaxis involves mast cell mediator release such as occurs with exercise or with cold temperature exposure, or from medications such as opioids or vancomycin. Idiopathic anaphylaxis involves mast cell activation (acutely elevated urine histamine or serum tryptase) and activated lymphocytes. Because anaphylaxis is a medical emergency, the drug of choice is epinephrine, not H1 antihistamines or H₂ receptor antagonists. DOI: 10.2500/aap.2019.40.4270 PMID: 31690393 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/34292177
1. Curr Opin Allergy Clin Immunol. 2021 Oct 1;21(5):426-434. doi: 10.1097/ACI.0000000000000768. Mast cell activation syndrome: is anaphylaxis part of the phenotype? A systematic review. Sabato V(1)(2), Michel M(3)(4)(5), Blank U(6), Ebo DG(1)(2), Vitte J(4)(5)(7). Author information: (1)University of Antwerp, Faculty of Medicine and Health Sciences, Department of Immunology, Allergology, Rheumatology and the Infla-Med Centre of Excellence. (2)AZ Jan Palfijn Gent, Department of Immunology and Allergology, Ghent, Belgium. (3)Laboratoire d'Immunologie, CHU de Nîmes, Nîmes. (4)Aix-Marseille Univ, IRD, APHM, MEPHI. (5)IHU Méditerranée Infection, Marseille. (6)Université de Paris, Centre de Recherche sur l'Inflammation, INSERM UMR1149, CNRS ERL8252, Faculté de Médecine site Bichat, Paris, France; Laboratoire d'Excellence Inflamex, Paris, France. (7)IDESP, INSERM UMR UA 11, Montpellier, France. PURPOSE OF REVIEW: Mast cell activation syndrome (MCAS) and anaphylaxis are the result of a spontaneous or triggered pathological degranulation of mast cells (MCs) and might have as substrate normal or pathological MCs (increased burden, aberrant MCs or both). RECENT FINDINGS: This review summarizes the most recent evidence on immunoglobulin E (IgE)-mediated and non IgE-mediated mechanisms underlying MC activation and degranulation and highlights the importance of standardized diagnostic criteria for MCAS. Application of these criteria implies that in most cases the clinical presentation of MCAS meets the diagnostic criteria for anaphylaxis. SUMMARY: Integrating clinical parameters and diagnostic test recognition and underlying clonal MC disease are of utmost importance for a patient-tailored approach. Hereditary alpha-tryptasemia can be encountered in context of anaphylaxis, MCAS and primary MC disorders. Copyright © 2021 Wolters Kluwer Health, Inc. All rights reserved. DOI: 10.1097/ACI.0000000000000768 PMID: 34292177 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/25240785
1. J Allergy Clin Immunol. 2015 Apr;135(4):1031-1043.e6. doi: 10.1016/j.jaci.2014.07.057. Epub 2014 Sep 18. Plasma contact system activation drives anaphylaxis in severe mast cell-mediated allergic reactions. Sala-Cunill A(1), Björkqvist J(2), Senter R(3), Guilarte M(4), Cardona V(4), Labrador M(4), Nickel KF(5), Butler L(5), Luengo O(4), Kumar P(2), Labberton L(2), Long A(6), Di Gennaro A(2), Kenne E(2), Jämsä A(2), Krieger T(6), Schlüter H(6), Fuchs T(5), Flohr S(7), Hassiepen U(7), Cumin F(7), McCrae K(8), Maas C(9), Stavrou E(10), Renné T(11). Author information: (1)Allergy Section, Internal Medicine Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Allergy Research Unit, Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain; Clinical Chemistry, Department of Molecular Medicine and Surgery, Karolinska Institutet and University Hospital, Stockholm, Sweden. (2)Clinical Chemistry, Department of Molecular Medicine and Surgery, Karolinska Institutet and University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden. (3)Clinical Chemistry, Department of Molecular Medicine and Surgery, Karolinska Institutet and University Hospital, Stockholm, Sweden; Department of Medicine, University of Padova, Padua, Italy. (4)Allergy Section, Internal Medicine Department, Hospital Universitari Vall d'Hebron, Barcelona, Spain; Allergy Research Unit, Institut de Recerca Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain. (5)Clinical Chemistry, Department of Molecular Medicine and Surgery, Karolinska Institutet and University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden; Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. (6)Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. (7)Novartis Institute for Biomedical Research, Novartis Campus, Basel, Switzerland. (8)Departments of Hematology and Medical Oncology and Cellular and Molecular Medicine, Cleveland Clinic, Cleveland, Ohio. (9)Department of Clinical Chemistry and Haematology, University Medical Center Utrecht, Utrecht, The Netherlands. (10)Department of Medicine, Hematology and Oncology Division, Case Western Reserve University and University Hospitals Case Medical Center, Cleveland, Ohio. (11)Clinical Chemistry, Department of Molecular Medicine and Surgery, Karolinska Institutet and University Hospital, Stockholm, Sweden; Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden; Institute for Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. Electronic address: [email protected]. BACKGROUND: Anaphylaxis is an acute, potentially lethal, multisystem syndrome resulting from the sudden release of mast cell-derived mediators into the circulation. OBJECTIVES AND METHODS: We report here that a plasma protease cascade, the factor XII-driven contact system, critically contributes to the pathogenesis of anaphylaxis in both murine models and human subjects. RESULTS: Deficiency in or pharmacologic inhibition of factor XII, plasma kallikrein, high-molecular-weight kininogen, or the bradykinin B2 receptor, but not the B1 receptor, largely attenuated allergen/IgE-mediated mast cell hyperresponsiveness in mice. Reconstitutions of factor XII null mice with human factor XII restored susceptibility for allergen/IgE-mediated hypotension. Activated mast cells systemically released heparin, which provided a negatively charged surface for factor XII autoactivation. Activated factor XII generates plasma kallikrein, which proteolyzes kininogen, leading to the liberation of bradykinin. We evaluated the contact system in patients with anaphylaxis. In all 10 plasma samples immunoblotting revealed activation of factor XII, plasma kallikrein, and kininogen during the acute phase of anaphylaxis but not at basal conditions or in healthy control subjects. The severity of anaphylaxis was associated with mast cell degranulation, increased plasma heparin levels, the intensity of contact system activation, and bradykinin formation. CONCLUSIONS: In summary, the data collectively show a role of the contact system in patients with anaphylaxis and support the hypothesis that targeting bradykinin generation and signaling provides a novel and alternative treatment strategy for anaphylactic attacks. Copyright © 2014 American Academy of Allergy, Asthma & Immunology. Published by Elsevier Inc. All rights reserved. DOI: 10.1016/j.jaci.2014.07.057 PMID: 25240785 [Indexed for MEDLINE]
http://www.ncbi.nlm.nih.gov/pubmed/31011586
1. J Immunol Res. 2019 Mar 18;2019:9542656. doi: 10.1155/2019/9542656. eCollection 2019. Anaphylactic Degranulation of Mast Cells: Focus on Compound Exocytosis. Klein O(1), Sagi-Eisenberg R(1). Author information: (1)Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel. Anaphylaxis is a notorious type 2 immune response which may result in a systemic response and lead to death. A precondition for the unfolding of the anaphylactic shock is the secretion of inflammatory mediators from mast cells in response to an allergen, mostly through activation of the cells via the IgE-dependent pathway. While mast cells are specialized secretory cells that can secrete through a variety of exocytic modes, the most predominant mode exerted by the mast cell during anaphylaxis is compound exocytosis-a specialized form of regulated exocytosis where secretory granules fuse to one another. Here, we review the modes of regulated exocytosis in the mast cell and focus on compound exocytosis. We review historical landmarks in the research of compound exocytosis in mast cells and the methods available for investigating compound exocytosis. We also review the molecular mechanisms reported to underlie compound exocytosis in mast cells and expand further with reviewing key findings from other cell types. Finally, we discuss the possible reasons for the mast cell to utilize compound exocytosis during anaphylaxis, the conflicting evidence in different mast cell models, and the open questions in the field which remain to be answered. DOI: 10.1155/2019/9542656 PMCID: PMC6442490 PMID: 31011586 [Indexed for MEDLINE]