51
|
Self WK, Holtzman DM. Emerging diagnostics and therapeutics for Alzheimer disease. Nat Med 2023; 29:2187-2199. [PMID: 37667136 DOI: 10.1038/s41591-023-02505-2] [Citation(s) in RCA: 144] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/18/2023] [Indexed: 09/06/2023]
Abstract
Alzheimer disease (AD) is the most common contributor to dementia in the world, but strategies that slow or prevent its clinical progression have largely remained elusive, until recently. This Review highlights the latest advances in biomarker technologies and therapeutic development to improve AD diagnosis and treatment. We review recent results that enable pathological staging of AD with neuroimaging and fluid-based biomarkers, with a particular emphasis on the role of amyloid, tau and neuroinflammation in disease pathogenesis. We discuss the lessons learned from randomized controlled trials, including some supporting the proposal that certain anti-amyloid antibodies slow cognitive decline during the mildly symptomatic phase of AD. In addition, we highlight evidence for newly identified therapeutic targets that may be able to modify AD pathogenesis and progression. Collectively, these recent discoveries-and the research directions that they open-have the potential to move AD clinical care toward disease-modifying treatment strategies with maximal benefits for patients.
Collapse
Affiliation(s)
- Wade K Self
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA
| | - David M Holtzman
- Department of Neurology, Hope Center for Neurological Disorders, Knight Alzheimer's Disease Research Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
52
|
Agarwal A, Gupta V, Brahmbhatt P, Desai A, Vibhute P, Joseph-Mathurin N, Bathla G. Amyloid-related Imaging Abnormalities in Alzheimer Disease Treated with Anti-Amyloid-β Therapy. Radiographics 2023; 43:e230009. [PMID: 37651273 DOI: 10.1148/rg.230009] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Alzheimer disease (AD) is the most common form of dementia worldwide. Treatment of AD has mainly been focused on symptomatic treatment until recently with the advent and approval of monoclonal antibody (MAB) immunotherapy. U.S. Food and Drug Administration-approved drugs such as aducanumab, as well as upcoming newer-generation drugs, have provided an exciting new therapy focused on reducing the amyloid plaque burden in AD. Although this new frontier has shown benefits for patients, it is not without complications, which are mainly neurologic. Increased use of MABs led to the discovery of amyloid-related imaging abnormalities (ARIA). ARIA has been further classified into two categories, ARIA-E and ARIA-H, representing edema and/or effusion and hemorrhage, respectively. ARIA is thought to be caused by increased vascular permeability following an inflammatory response, leading to the extravasation of blood products and proteinaceous fluid. Patients with ARIA may present with headaches, but they are usually asymptomatic and ARIA is only diagnosable at MRI; it is essential for the radiologist to recognize and monitor ARIA. Increased incidence and investigation into this concern have led to the creation of grading scales and monitoring guidelines to diagnose and guide treatment using MABs. Cerebral amyloid angiopathy has an identical pathogenesis to that of ARIA and is its closest differential diagnosis, with imaging findings being the same for both entities and only a history of MAB administration allowing differentiation. The authors discuss the use of MABs for treating AD, expand on ARIA and its consequences, and describe how to identify and grade ARIA to guide treatment properly. ©RSNA, 2023 Quiz questions for this article are available through the Online Learning Center See the invited commentary by Yu in this issue.
Collapse
Affiliation(s)
- Amit Agarwal
- From the Departments of Radiology (A.A., V.G., P.B., A.D.) and Neuroradiology (P.V.), Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL 32224; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Mo (N.J.M.); and Department of Radiology, Mayo Clinic, Rochester, Minn (G.B.)
| | - Vivek Gupta
- From the Departments of Radiology (A.A., V.G., P.B., A.D.) and Neuroradiology (P.V.), Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL 32224; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Mo (N.J.M.); and Department of Radiology, Mayo Clinic, Rochester, Minn (G.B.)
| | - Pavan Brahmbhatt
- From the Departments of Radiology (A.A., V.G., P.B., A.D.) and Neuroradiology (P.V.), Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL 32224; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Mo (N.J.M.); and Department of Radiology, Mayo Clinic, Rochester, Minn (G.B.)
| | - Amit Desai
- From the Departments of Radiology (A.A., V.G., P.B., A.D.) and Neuroradiology (P.V.), Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL 32224; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Mo (N.J.M.); and Department of Radiology, Mayo Clinic, Rochester, Minn (G.B.)
| | - Prasanna Vibhute
- From the Departments of Radiology (A.A., V.G., P.B., A.D.) and Neuroradiology (P.V.), Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL 32224; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Mo (N.J.M.); and Department of Radiology, Mayo Clinic, Rochester, Minn (G.B.)
| | - Nelly Joseph-Mathurin
- From the Departments of Radiology (A.A., V.G., P.B., A.D.) and Neuroradiology (P.V.), Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL 32224; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Mo (N.J.M.); and Department of Radiology, Mayo Clinic, Rochester, Minn (G.B.)
| | - Girish Bathla
- From the Departments of Radiology (A.A., V.G., P.B., A.D.) and Neuroradiology (P.V.), Mayo Clinic, 4500 San Pablo Rd, Jacksonville, FL 32224; Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Mo (N.J.M.); and Department of Radiology, Mayo Clinic, Rochester, Minn (G.B.)
| |
Collapse
|
53
|
Theodorou A, Palaiodimou L, Papagiannopoulou G, Kargiotis O, Psychogios K, Safouris A, Bakola E, Chondrogianni M, Kotsali-Peteinelli V, Melanis K, Tsibonakis A, Andreadou E, Vasilopoulou S, Lachanis S, Velonakis G, Tzavellas E, Tzartos JS, Voumvourakis K, Paraskevas GP, Tsivgoulis G. Clinical Characteristics, Neuroimaging Markers, and Outcomes in Patients with Cerebral Amyloid Angiopathy: A Prospective Cohort Study. J Clin Med 2023; 12:5591. [PMID: 37685658 PMCID: PMC10488273 DOI: 10.3390/jcm12175591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/23/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Background and purpose: Sporadic cerebral amyloid angiopathy (CAA) is a small vessel disease, resulting from progressive amyloid-β deposition in the media/adventitia of cortical and leptomeningeal arterioles. We sought to assess the prevalence of baseline characteristics, clinical and radiological findings, as well as outcomes among patients with CAA, in the largest study to date conducted in Greece. Methods: Sixty-eight patients fulfilling the Boston Criteria v1.5 for probable/possible CAA were enrolled and followed for at least twelve months. Magnetic Resonance Imaging was used to assess specific neuroimaging markers. Data regarding cerebrospinal fluid biomarker profile and Apolipoprotein-E genotype were collected. Multiple logistic regression analyses were performed to identify predictors of clinical phenotypes. Cox-proportional hazard regression models were used to calculate associations with the risk of recurrent intracerebral hemorrhage (ICH). Results: Focal neurological deficits (75%), cognitive decline (57%), and transient focal neurological episodes (TFNEs; 21%) were the most common clinical manifestations. Hemorrhagic lesions, including lobar cerebral microbleeds (CMBs; 93%), cortical superficial siderosis (cSS; 48%), and lobar ICH (43%) were the most prevalent neuroimaging findings. cSS was independently associated with the likelihood of TFNEs at presentation (OR: 4.504, 95%CI:1.258-19.088), while multiple (>10) lobar CMBs were independently associated with cognitive decline at presentation (OR:5.418, 95%CI:1.316-28.497). cSS emerged as the only risk factor of recurrent ICH (HR:4.238, 95%CI:1.509-11.900) during a median follow-up of 20 months. Conclusions: cSS was independently associated with TFNEs at presentation and ICH recurrence at follow-up, while a higher burden of lobar CMBs with cognitive decline at baseline. These findings highlight the prognostic value of neuroimaging markers, which may influence clinical decision-making.
Collapse
Affiliation(s)
- Aikaterini Theodorou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Lina Palaiodimou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Georgia Papagiannopoulou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Odysseas Kargiotis
- Stroke Unit, Metropolitan Hospital, 18547 Piraeus, Greece; (O.K.); (K.P.); (A.S.)
| | - Klearchos Psychogios
- Stroke Unit, Metropolitan Hospital, 18547 Piraeus, Greece; (O.K.); (K.P.); (A.S.)
| | - Apostolos Safouris
- Stroke Unit, Metropolitan Hospital, 18547 Piraeus, Greece; (O.K.); (K.P.); (A.S.)
| | - Eleni Bakola
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Maria Chondrogianni
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Vasiliki Kotsali-Peteinelli
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Konstantinos Melanis
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Athanasios Tsibonakis
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Elissavet Andreadou
- First Department of Neurology, “Eginition” Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (E.A.); (S.V.)
| | - Sofia Vasilopoulou
- First Department of Neurology, “Eginition” Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece; (E.A.); (S.V.)
| | - Stefanos Lachanis
- Iatropolis Magnetic Resonance Diagnostic Centre, 15231 Athens, Greece;
| | - Georgios Velonakis
- Second Department of Radiology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece;
| | - Elias Tzavellas
- First Department of Psychiatry, “Aiginition” Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece;
| | - John S. Tzartos
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Konstantinos Voumvourakis
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Georgios P. Paraskevas
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
| | - Georgios Tsivgoulis
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (A.T.); (L.P.); (G.P.); (E.B.); (M.C.); (V.K.-P.); (K.M.); (A.T.); (J.S.T.); (K.V.); (G.P.P.)
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
54
|
Theodorou A, Tsibonakis A, Pateras IS, Kaloudi G, Bakola E, Chondrogianni M, Andreadou E, Panayiotides IG, Tsivgoulis G. Multiple cerebral microinfarcts: an uncommon presentation of Cerebral Amyloid Angiopathy-related inflammation. Neurol Res Pract 2023; 5:28. [PMID: 37344910 DOI: 10.1186/s42466-023-00253-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/26/2023] [Indexed: 06/23/2023] Open
Abstract
BACKGROUND Cerebral Amyloid Angiopathy-related inflammation (CAA-ri) is a distinct but rare subset of CAA. The greater availability of high resolution Magnetic Resonance Imaging (MRI) has currently allowed the increasing recognition and diagnosis of this entity, without the risk of a brain biopsy. However, in rare cases with typical clinical characteristics but uncommon neuroimaging findings at presentation, the brain-biopsy is required for an early and reliable diagnosis. CASE DESCRIPTION A 71-year-old man with arterial hypertension presented due to 1-week history of headache, vomiting, disorientation and impaired consciousness. Brain MRI revealed multiple acute cortical/subcortical microinfarcts, scarce microbleeds, extensive right parietooccipital and left frontotemporal leptomeningeal enhancement. After an extensive diagnostic work-up, excluding infectious, neoplastic and autoimmune etiologies, the patient underwent brain-biopsy. Histology disclosed amyloid deposition in an arteriolar wall and the patient fulfilled diagnostic criteria for probable CAA-ri with supporting pathology. He received intravenous methylprednisolone, followed by oral tapering with steroids showing clinical and radiological improvement with complete resolution of gadolinium enhancement. Follow-up MRI revealed an increase of cerebral microbleeds and the patient fulfilled CAA-ri neuroimaging criteria. CONCLUSIONS This case highlights the importance of continuous vigilance from clinical neurologists to detect CAA-ri diagnosis and the diagnostic value of brain-biopsy in CAA-ri patients with atypical neuroimaging presentation, such as acute microinfarcts. The early diagnosis and the prompt treatment initiation can improve the prognosis and the evolution of this rare disorder.
Collapse
Affiliation(s)
- Aikaterini Theodorou
- Second Department of Neurology, School of Medicine, "Attikon" University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Athanasios Tsibonakis
- Second Department of Neurology, School of Medicine, "Attikon" University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis S Pateras
- Second Department of Pathology, School of Medicine, "Attikon" University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgia Kaloudi
- Second Department of Neurology, School of Medicine, "Attikon" University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleni Bakola
- Second Department of Neurology, School of Medicine, "Attikon" University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Chondrogianni
- Second Department of Neurology, School of Medicine, "Attikon" University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Elissavet Andreadou
- First Department of Neurology, School of Medicine, "Eginition" University Hospital, National and Kapodistiran University of Athens, Athens, Greece
| | - Ioannis G Panayiotides
- Second Department of Pathology, School of Medicine, "Attikon" University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgios Tsivgoulis
- Second Department of Neurology, School of Medicine, "Attikon" University Hospital, National and Kapodistrian University of Athens, Athens, Greece.
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
55
|
Hansen N, Juhl AL, Grenzer IM, Teegen B, Wiltfang J, Fitzner D. Cerebrospinal fluid biomarkers in psychiatric autoimmune encephalitis: a retrospective cohort study. Front Psychiatry 2023; 14:1165153. [PMID: 37363167 PMCID: PMC10287966 DOI: 10.3389/fpsyt.2023.1165153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 05/16/2023] [Indexed: 06/28/2023] Open
Abstract
Background Psychiatric autoimmune encephalitis (pAE) is a growing field of interest in diagnosis and therapy in psychiatric hospitals and institutions. This study investigates the relevant extent to which there are potential biomarkers in cerebrospinal fluid (CSF) that can differentiate against a cohort with neurodegenerative disease. Methods We included in this study a total of 27 patients with possible and definite psychiatric autoimmune encephalitis and compared with a cohort with CSF-based AD (n = 27) different biomarkers in CSF such as lactate, cell count, % lymphocytes, % monocytes, total protein content, albumin, immunoglobulins G (IgG), M (IgM) and A (IgA), CSF/serum albumin ratio, CSF/serum IgG ratio, CSF/serum IgA ratio, intrathecal IgG synthesis, blood-brain barrier disruption, specific antibody synthesis for measles, rubella, herpes simplex virus, varicella zoster virus, Ebstein-Barr virus and cytomegalovirus, total tau protein (t-tau), phosphorylated tau protein 181 (p-tau181), amyloid beta 42 (Aß42), amyloid beta 40 (Aß40) and the amyloid beta 42/ amyloid beta 40 (Aß42/40) ratio. Results The p-tau 181 was elevated above cut-off values in both possible pAE and AD. However, in definitive pAE, p-tau181 levels were not elevated. When elevated p-tau181 levels in possible AE were compared with those in AD, we found relevant differences, such as a relative increase in p-tau181 in AD patients. Elevated p-tau181 levels were detected in possible psychiatric AEs with IgLON5, glycine, recoverin, titin, and nonspecific neuropil antibodies in serum and IgLON5, titin, Yo, and nonspecific neuropil autoantibodies in CSF. In addition, we detected elevated levels of p-tau181 and IgLON5 autoantibodies in serum and CSF, and Yo autoantibodies in CSF in patients with definitive pAE. Interestingly, we observed a higher CSF/serum IgM ratio in possible and definitive pAE than in AD patients. Conclusion Our results suggest that neuroaxonal brain damage may occur in specific psychiatric AEs associated with IgLON5, glycine, recoverin, and titin autoantibodies. Further research should focus on the CSF/serum IgM ratio as an early marker of autoantibody production in pAE compared to AD as a potential biomarker for differential diagnosis.
Collapse
Affiliation(s)
- Niels Hansen
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Aaron Levin Juhl
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Insa Maria Grenzer
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
| | - Bianca Teegen
- Clinical Immunological Laboratory Prof. Stöcker, Groß Grönau, Germany
| | - Jens Wiltfang
- Department of Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- German Center for Neurodegenerative Diseases (DZNE), Göttingen, Germany
- Neurosciences and Signaling Group, Institute of Biomedicine (iBiMED), Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Dirk Fitzner
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
56
|
de Souza A, Tasker K. Inflammatory Cerebral Amyloid Angiopathy: A Broad Clinical Spectrum. J Clin Neurol 2023; 19:230-241. [PMID: 37151140 PMCID: PMC10169922 DOI: 10.3988/jcn.2022.0493] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 03/28/2023] [Accepted: 03/31/2023] [Indexed: 05/09/2023] Open
Abstract
Cerebral amyloid angiopathy (CAA) is a common central nervous system (CNS) vasculopathy, which in some cases is associated with subacute encephalopathy, seizures, headaches, or strokes due to vascular inflammation directed against vascular amyloid accumulation. The pathological subtypes of inflammatory CAA include CAA-related inflammation (CAAri) with mostly perivascular lymphocytic infiltrates, or amyloid-beta (Aβ)-related angiitis (ABRA) with transmural granulomatous inflammation. CAAri and ABRA probably represent part of the spectrum of CNS vasculopathies, intermediate between CAA and primary CNS vasculitis, and they are closely related to Aβ-related imaging abnormalities and other manifestations of an inflammatory response directed against Aβ in the leptomeninges and cerebral parenchyma. As treatment strategies in Alzheimer's disease shift toward potentially effective antiamyloid immunotherapy, the incidence rate of inflammatory CAA (which is probably an underrecognized condition) is likely to increase. Its clinical features are varied and include subacute encephalopathy, behavioral symptoms, headaches, seizures, and focal neurological deficits, which necessitate a high degree of suspicion for this disorder that often responds to treatment. The recent definition of the typical clinical and radiological syndrome has increased its recognition and may eliminate the need for invasive histological sampling in at least some affected patients. Here we review the pathophysiology, clinical spectrum, and approach to diagnosis, and discuss illustrative cases that highlight the wide range of clinical presentations.
Collapse
Affiliation(s)
- Aaron de Souza
- Department of Medicine, Launceston General Hospital, Launceston, Australia
- Faculty of Medicine, Launceston Clinical School, University of Tasmania, Launceston, Australia.
| | - Kate Tasker
- Department of Medicine, Launceston General Hospital, Launceston, Australia
| |
Collapse
|
57
|
Castellani RJ, Shanes E, McCord M, Reish NJ, Flanagan ME, Mesulam MM, Jamshidi P. Neuropathology of Anti-Amyloid-β Immunotherapy: A Case Report. J Alzheimers Dis 2023; 93:803-813. [PMID: 37125554 DOI: 10.3233/jad-221305] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Host responses to anti-amyloid-β (Aβ) antibody therapy are evident in neuroimaging changes and clinical symptoms in a subset of clinical trial subjects receiving such therapy. The pathological basis for the imaging changes and clinical symptoms is not known, nor is the precise mechanism of Aβ clearing. We report the autopsy findings in a 65-year-old woman who received three open label infusions of the experimental anti-Aβ drug lecanemab over about one month. Four days after the last infusion, she was treated with tissue plasminogen activator for acute stroke symptoms and died several days later with multifocal hemorrhage. Neuropathological examination demonstrated histiocytic vasculitis involving blood vessels with cerebral amyloid angiopathy. Fragmentation and phagocytosis of vascular Aβ were present throughout the cerebral cortex. Phagocytosis of parenchymal Aβ plaques was noted. Changes suggestive of Aβ and phosphorylated tau "clearing" were also noted. The findings overall suggest that anti-Aβ treatment stimulated a host response to Aβ, i.e., target engagement. The findings also provide evidence that amyloid-related imaging abnormalities might be indicative of an Aβ phagocytic syndrome within cerebral vasculature and parenchymal brain tissue in some cases.
Collapse
Affiliation(s)
- Rudolph J Castellani
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Elisheva Shanes
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Matthew McCord
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Nicholas J Reish
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Margaret E Flanagan
- Department of Pathology, University of Texas, San Antonio, San Antonio, TX, USA
| | - M-Marsel Mesulam
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Pouya Jamshidi
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
58
|
Sakai K, Noguchi-Shinohara M, Tanaka H, Ikeda T, Hamaguchi T, Kakita A, Yamada M, Ono K. Cerebrospinal Fluid Biomarkers and Amyloid-β Elimination from the Brain in Cerebral Amyloid Angiopathy-Related Inflammation. J Alzheimers Dis 2023; 91:1173-1183. [PMID: 36565118 DOI: 10.3233/jad-220838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Cerebrospinal fluid (CSF) biomarkers in patients with cerebral amyloid angiopathy-related inflammation (CAA-ri) have demonstrated inconsistent results. OBJECTIVE We investigated the relationship between CSF amyloid-β protein (Aβ) and vascular pathological findings to elucidate the mechanisms of Aβ elimination from the brain in CAA-ri. METHODS We examined Aβ40 and Aβ42 levels in CSF samples in 15 patients with CAA-ri and 15 patients with Alzheimer's disease and cerebral amyloid angiopathy (AD-CAA) using ELISA as a cross-sectional study. Furthermore, we pathologically examined Aβ40 and Aβ42 depositions on the leptomeningeal blood vessels (arteries, arterioles, and veins) using brain biopsy samples from six patients with acute CAA-ri and brain tissues of two autopsied patients with CAA-ri. RESULTS The median Aβ40 and Aβ42 levels in the CSF showed no significant difference between pre-treatment CAA-ri (Aβ40, 6837 pg/ml; Aβ42, 324 pg/ml) and AD-CAA (Aβ40, 7669 pg/ml, p = 0.345; Aβ42, 355 pg/ml, p = 0.760). Aβ40 and Aβ42 levels in patients with post-treatment CAA-ri (Aβ40, 1770 pg/ml, p = 0.056; Aβ42, 167 pg/ml, p = 0.006) were lower than those in patients with pre-treatment CAA-ri. Regarding Aβ40 and Aβ42 positive arteries, acute CAA-ri cases showed a higher frequency of partially Aβ-deposited blood vessels than postmortem CAA-ri cases (Aβ40, 20.8% versus 3.9%, p = 0.0714; Aβ42, 27.4% versus 2.0%, p = 0.0714, respectively). CONCLUSION Lower levels of CSF Aβ40 and Aβ42 could be biomarkers for the cessation of inflammation in CAA-ri reflecting the recovery of the intramural periarterial drainage pathway and vascular function.
Collapse
Affiliation(s)
- Kenji Sakai
- Department of Neurology, Joetsu General Hospital, Joetsu, Japan.,Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Moeko Noguchi-Shinohara
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan.,Department of Preemptive Medicine for Dementia, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Hidetomo Tanaka
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Tokuhei Ikeda
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Tsuyoshi Hamaguchi
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan.,Department of Neurology, Kanazawa Medical University, Uchinada, Japan
| | - Akiyoshi Kakita
- Department of Pathology, Brain Research Institute, Niigata University, Niigata, Japan
| | - Masahito Yamada
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan.,Department of Internal Medicine, Kudanzaka Hospital, Tokyo, Japan
| | - Kenjiro Ono
- Department of Neurology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| |
Collapse
|
59
|
Li D, Qin W, Guo Y, Xia M, Li S, Zhang J, Zang W. Clinical, laboratory, and radiological features of cerebral amyloid angiopathy-related inflammation (CAA-ri): retrospective, observational experience of a single centre. Neurol Sci 2023; 44:631-638. [PMID: 36207650 DOI: 10.1007/s10072-022-06436-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND Cerebral amyloid angiopathy-related inflammation (CAA-ri) is a subtype of CAA with an inflammatory response to the vascular β-amyloid deposits. Reliable and non-invasive clinical diagnostic methods may allow patients to avoid the side effects of brain biopsy. OBJECTIVE In this observational study, we retrospectively analyzed the clinical, laboratory, radiological features, treatment, and outcome of patients diagnosed with CAA-ri. The main purpose is to enhance knowledge of CAA-ri and to avoid misdiagnosis. METHODS We described 15 consecutive patients with probable or possible CAA-ri at Henan Provincial People's Hospital according to a validation study of proposed criteria for the diagnosis of CAA-ri. The clinical features, imaging, laboratory findings, and treatment which included the response to immunotherapy were revealed in the study. RESULTS The median age of 15 patients was 67.0 years (range 48.0-90.0 years), and the male-to-female ratio was 7: 8. In our study, the most common clinical manifestations were cognitive decline (7/15, 46.7%), focal neurologic deficit (6/15, 40.0%), and headache (5/15, 33.3%). In terms of imaging results, white matter hyperintensity (WMH) lesions were rarely seen in the cerebellum and brainstem, while no hemorrhagic lesion was observed in the brainstem of all 15 patients. In addition, 12 patients (80.0%) showed improvement or stability for the clinical and radiological outcomes after immunotherapy. CONCLUSION CAA-ri should be considered as a differential diagnosis when brain MRI shows typical features in the elderly. Once the diagnosis is established, immunotherapy should be initiated as early as possible to promote neurological function recovery and reduce recurrence.
Collapse
Affiliation(s)
- Dan Li
- Department of Neurology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No. 7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Weiwei Qin
- Department of Neurology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No. 7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Yang Guo
- Department of Neurosurgery, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No. 7 Weiwu Road, 450003, Zhengzhou, China
| | - Mingrong Xia
- Department of Neurology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No. 7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Siyuan Li
- Department of Neurology, Dengfeng People's Hospital, Dengfeng, 452470, Henan, China
| | - Jiewen Zhang
- Department of Neurology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No. 7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China
| | - Weizhou Zang
- Department of Neurology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, No. 7 Weiwu Road, Jinshui District, Zhengzhou, 450003, Henan, China.
| |
Collapse
|
60
|
Zedde M, Pascarella R, Piazza F. CAA-ri and ARIA: Two Faces of the Same Coin? AJNR Am J Neuroradiol 2023; 44:E13-E14. [PMID: 36635054 PMCID: PMC9891329 DOI: 10.3174/ajnr.a7759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- M Zedde
- Neurology Unit, Stroke UnitAzienda Unità Sanitaria Locale-IRCCS di Reggio EmiliaReggio Emilia, Italy
| | - R Pascarella
- Neuroradiology Unit, iCAβ International NetworkAzienda Unità Sanitaria Locale-IRCCS di Reggio EmiliaReggio Emilia, Italy
| | - F Piazza
- CAA and AD Translational Research and Biomarkers Lab, School of MedicineUniversity of Milano-BicoccaMonza, Italy
| |
Collapse
|
61
|
Tabaee Damavandi P, Storti B, Fabin N, Bianchi E, Ferrarese C, DiFrancesco JC. Epilepsy in cerebral amyloid angiopathy: an observational retrospective study of a large population. Epilepsia 2023; 64:500-510. [PMID: 36515439 DOI: 10.1111/epi.17489] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 12/02/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Cerebral amyloid angiopathy (CAA) is a major cause of spontaneous intracranial hemorrhage in older adults. Epilepsy represents a possible sequela of the disease. To date, studies on epilepsy in CAA are lacking, and the few data available mainly focus on CAA-related inflammation (CAA-ri), the inflammatory form of the disease. METHODS In this retrospective observational study, we consecutively recruited CAA patients observed over a time span of 10 years, collecting demographic, clinical, and instrumental data. Significant baseline characteristics were evaluated as potential risk factors for the development of epilepsy in the CAA population, and in the subgroups of CAA-ri and CAA without inflammatory reaction (CAA-nri). The effect of potential risk factors for epilepsy was measured as odds ratio with 95% confidence interval. RESULTS Within 96 recruited CAA cases, 33 (34.4%) developed epilepsy during follow-up (median = 13.5 months). The prevalent type of seizure was focal (81.3%); 12.1% of the epileptic patients presented status epilepticus, and 6.1% developed drug-resistant epilepsy. Electroencephalographic traces revealed slow and epileptic discharge activity in the majority of epileptic patients, but also in those without epilepsy. The presence of focal or disseminated cortical superficial siderosis (cSS) was associated with an increased risk of epilepsy in the CAA-nri group, and the association with CAA-ri and epilepsy was present in the overall population. SIGNIFICANCE Epilepsy is a common manifestation during the course of CAA, where CAA-ri and cSS represent predisposing factors for the development of seizures. These data suggest the importance of a deep characterization of CAA patients, to better select those more prone to develop epilepsy.
Collapse
Affiliation(s)
- Payam Tabaee Damavandi
- Department of Neurology, ASST San Gerardo Hospital, School of Medicine and Surgery and Milan Center for Neuroscience, University of Milan-Bicocca, Monza, Italy
| | - Benedetta Storti
- Department of Neurology, ASST San Gerardo Hospital, School of Medicine and Surgery and Milan Center for Neuroscience, University of Milan-Bicocca, Monza, Italy
| | - Natalia Fabin
- Laboratory of Epidemiological and Clinical Cardiology, Department of Experimental, Diagnostic, and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Elisa Bianchi
- Neurological Disorders, Mario Negri Institute of Pharmacological Research, Scientific Institute for Research and Health Care, Milan, Italy
| | - Carlo Ferrarese
- Department of Neurology, ASST San Gerardo Hospital, School of Medicine and Surgery and Milan Center for Neuroscience, University of Milan-Bicocca, Monza, Italy
| | - Jacopo C DiFrancesco
- Department of Neurology, ASST San Gerardo Hospital, School of Medicine and Surgery and Milan Center for Neuroscience, University of Milan-Bicocca, Monza, Italy
| |
Collapse
|
62
|
Ohashi SN, DeLong JH, Kozberg MG, Mazur-Hart DJ, van Veluw SJ, Alkayed NJ, Sansing LH. Role of Inflammatory Processes in Hemorrhagic Stroke. Stroke 2023; 54:605-619. [PMID: 36601948 DOI: 10.1161/strokeaha.122.037155] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Hemorrhagic stroke is the deadliest form of stroke and includes the subtypes of intracerebral hemorrhage and subarachnoid hemorrhage. A common cause of hemorrhagic stroke in older individuals is cerebral amyloid angiopathy. Intracerebral hemorrhage and subarachnoid hemorrhage both lead to the rapid collection of blood in the central nervous system and generate inflammatory immune responses that involve both brain resident and infiltrating immune cells. These responses are complex and can contribute to both tissue recovery and tissue injury. Despite the interconnectedness of these major subtypes of hemorrhagic stroke, few reviews have discussed them collectively. The present review provides an update on inflammatory processes that occur in response to intracerebral hemorrhage and subarachnoid hemorrhage, and the role of inflammation in the pathophysiology of cerebral amyloid angiopathy-related hemorrhage. The goal is to highlight inflammatory processes that underlie disease pathology and recovery. We aim to discuss recent advances in our understanding of these conditions and identify gaps in knowledge with the potential to develop effective therapeutic strategies.
Collapse
Affiliation(s)
- Sarah N Ohashi
- Department of Neurology (S.N.O., J.H.D., L.H.S.), Yale School of Medicine, New Haven, CT
- Department of Immunobiology (S.N.O., J.H.D., L.H.S.), Yale School of Medicine, New Haven, CT
| | - Jonathan H DeLong
- Department of Neurology (S.N.O., J.H.D., L.H.S.), Yale School of Medicine, New Haven, CT
- Department of Immunobiology (S.N.O., J.H.D., L.H.S.), Yale School of Medicine, New Haven, CT
| | - Mariel G Kozberg
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital/ Harvard Medical School, Boston (M.G.K., S.J.v.V.)
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown (M.G.K., S.J.v.V.)
| | - David J Mazur-Hart
- Department of Neurological Surgery (D.J.M.-H.), Oregon Health and Science University (OHSU), Portland
| | - Susanne J van Veluw
- J. Philip Kistler Stroke Research Center, Department of Neurology, Massachusetts General Hospital/ Harvard Medical School, Boston (M.G.K., S.J.v.V.)
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Charlestown (M.G.K., S.J.v.V.)
| | - Nabil J Alkayed
- Department of Anesthesiology & Perioperative Medicine and Knight Cardiovascular Institute (N.J.A.), Oregon Health and Science University (OHSU), Portland
| | - Lauren H Sansing
- Department of Neurology (S.N.O., J.H.D., L.H.S.), Yale School of Medicine, New Haven, CT
- Department of Immunobiology (S.N.O., J.H.D., L.H.S.), Yale School of Medicine, New Haven, CT
| |
Collapse
|
63
|
Yang JY, Chu YT, Tsai HH, Jeng JS. Amyloid and tau PET in cerebral amyloid angiopathy-related inflammation two case reports and literature review. Front Neurol 2023; 14:1153305. [PMID: 37188315 PMCID: PMC10175602 DOI: 10.3389/fneur.2023.1153305] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Background Cerebral amyloid angiopathy-related inflammation (CAA-ri) is a clinical syndrome characterized by MRI findings of amyloid-related imaging abnormalities-edema (ARIA-E) suggestive of autoimmune and inflammatory reaction and hemorrhagic evidence of cerebral amyloid angiopathy. The longitudinal variation of amyloid PET and its imaging association with CAA-ri are undetermined. Moreover, tau PET in CAA-ri has been rarely investigated. Method We retrospectively described two cases of CAA-ri. We provided the temporal change of amyloid and tau PET in the first case, and the cross-sectional finding of amyloid and tau PET in the second case. We also performed a literature review of the imaging features of amyloid PET in reported cases of CAA-ri. Results In the first case, an 88-year-old male presented with progressive consciousness and gait disturbances over 2 months. MRI showed disseminated cortical superficial siderosis. Amyloid PET prior to and after the CAA-ri revealed focally decreased amyloid load in the region of ARIA-E. In the second case, a 72-year-old male was initially suspected to have central nervous system cryptococcosis but later diagnosed with CAA-ri because of the characteristic MRI features and good response to corticosteroid treatment; a subsequent amyloid scan revealed positive amyloid deposition of the brain. Neither case suggested an association between the region of ARIA-E and higher amyloid uptake on PET before or after onset of CAA-ri. Our literature review revealed variable findings related to amyloid burden in post-inflammatory regions in previously reported CAA-ri cases with available amyloid PET. Our case is the first report of longitudinal changes on amyloid PET and show focal decreases in amyloid load after the inflammatory process. Conclusion This case series highlights the need to better explore the potential of longitudinal amyloid PET in the understanding of the mechanisms of CAA-ri.
Collapse
Affiliation(s)
- Jhih-Yong Yang
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yung-Tsai Chu
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Hsin-Hsi Tsai
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
- Department of Neurology, National Taiwan University Hospital Bei-Hu Branch, Taipei, Taiwan
- *Correspondence: Hsin-Hsi Tsai
| | - Jiann-Shing Jeng
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
64
|
Cummings J, Apostolova L, Rabinovici GD, Atri A, Aisen P, Greenberg S, Hendrix S, Selkoe D, Weiner M, Petersen RC, Salloway S. Lecanemab: Appropriate Use Recommendations. J Prev Alzheimers Dis 2023; 10:362-377. [PMID: 37357276 PMCID: PMC10313141 DOI: 10.14283/jpad.2023.30] [Citation(s) in RCA: 220] [Impact Index Per Article: 110.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Abstract
Lecanemab (Leqembi®) is approved in the United States for the treatment of Alzheimer's disease (AD) to be initiated in early AD (mild cognitive impairment [MCI] due to AD or mild AD dementia) with confirmed brain amyloid pathology. Appropriate Use Recommendations (AURs) are intended to help guide the introduction of new therapies into real-world clinical practice. Community dwelling patients with AD differ from those participating in clinical trials. Administration of lecanemab at clinical trial sites by individuals experienced with monoclonal antibody therapy also differs from the community clinic-based administration of lecanemab. These AURs use clinical trial data as well as research and care information regarding AD to help clinicians administer lecanemab with optimal safety and opportunity for effectiveness. Safety and efficacy of lecanemab are known only for patients like those participating in the phase 2 and phase 3 lecanemab trials, and these AURs adhere closely to the inclusion and exclusion criteria of the trials. Adverse events may occur with lecanemab including amyloid related imaging abnormalities (ARIA) and infusion reactions. Monitoring guidelines for these events are detailed in this AUR. Most ARIA with lecanemab is asymptomatic, but a few cases are serious or, very rarely, fatal. Microhemorrhages and rare macrohemorrhages may occur in patients receiving lecanemab. Anticoagulation increases the risk of hemorrhage, and the AUR recommends that patients requiring anticoagulants not receive lecanemab until more data regarding this interaction are available. Patients who are apolipoprotein E ε4 (APOE4) gene carriers, especially APOE4 homozygotes, are at higher risk for ARIA, and the AUR recommends APOE genotyping to better inform risk discussions with patients who are lecanemab candidates. Clinician and institutional preparedness are mandatory for use of lecanemab, and protocols for management of serious events should be developed and implemented. Communication between clinicians and therapy candidates or those on therapy is a key element of good clinical practice for the use of lecanemab. Patients and their care partners must understand the potential benefits, the potential harms, and the monitoring requirements for treatment with this agent. Culture-specific communication and building of trust between clinicians and patients are the foundation for successful use of lecanemab.
Collapse
Affiliation(s)
- J Cummings
- Jeffrey Cummings, MD, ScD, 1380 Opal Valley Street, Henderson, NV 89052, USA, , T: 702-902-3939
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Theodorou A, Palaiodimou L, Malhotra K, Zompola C, Katsanos AH, Shoamanesh A, Boviatsis E, Dardiotis E, Spilioti M, Sacco S, Werring DJ, Cordonnier C, Alexandrov AV, Paraskevas GP, Tsivgoulis G. Clinical, Neuroimaging, and Genetic Markers in Cerebral Amyloid Angiopathy-Related Inflammation: A Systematic Review and Meta-Analysis. Stroke 2023; 54:178-188. [PMID: 36453271 DOI: 10.1161/strokeaha.122.040671] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
BACKGROUND There are limited data regarding the prevalence of distinct clinical, neuroimaging and genetic markers among patients diagnosed with cerebral amyloid angiopathy-related inflammation (CAA-ri). We sought to determine the prevalence of clinical, radiological, genetic and cerebrospinal fluid biomarker findings in patients with CAA-ri. METHODS A systematic review and meta-analysis of published studies including patients with CAA-ri was conducted to determine the prevalence of clinical, neuroimaging, genetic and cerebrospinal fluid biomarker findings. Subgroup analyses were performed based on (1) prospective or retrospective study design and (2) CAA-ri diagnosis with or without available biopsy. We pooled the prevalence rates using random-effects models and assessed the heterogeneity using Cochran-Q and I2-statistics. RESULTS We identified 4 prospective and 17 retrospective cohort studies comprising 378 patients with CAA-ri (mean age, 71.5 years; women, 52%). The pooled prevalence rates were as follows: cognitive decline at presentation 70% ([95% CI, 54%-84%]; I2=82%), focal neurological deficits 55% ([95% CI, 40%-70%]; I2=82%), encephalopathy 54% ([95% CI, 39%-68%]; I2=43%), seizures 37% ([95% CI, 27%-49%]; I2=65%), headache 31% ([95% CI, 22%-42%]; I2=58%), T2/fluid-attenuated inversion recovery-hyperintense white matter lesions 98% ([95% CI, 93%-100%]; I2=44%), lobar cerebral microbleeds 96% ([95% CI, 92%-99%]; I2=25%), gadolinium enhancing lesions 54% ([95% CI, 42%-66%]; I2=62%), cortical superficial siderosis 51% ([95% CI, 34%-68%]; I2=77%) and lobar macrohemorrhage 40% ([95% CI, 11%-73%]; I2=88%). The prevalence rate of the ApoE (Apolipoprotein E) ε4/ε4 genotype was 34% ([95% CI, 17%-53%]; I2=76%). Subgroup analyses demonstrated no differences in these prevalence rates based on study design and diagnostic strategy. CONCLUSIONS Cognitive decline was the most common clinical feature. Hyperintense T2/fluid-attenuated inversion recovery white matter lesions and lobar cerebral microbleeds were by far the most prevalent neuroimaging findings. Thirty-four percent of patients with CAA-ri have homozygous ApoE ε4/ε4 genotype and scarce data exist regarding the cerebrospinal fluid biomarkers and its significance in these patients.
Collapse
Affiliation(s)
- Aikaterini Theodorou
- Second Department of Neurology (A.T., L.P., C.Z., G.P.P., G.T.), National & Kapodistrian University of Athens, "Attikon" University Hospital, Greece
| | - Lina Palaiodimou
- Second Department of Neurology (A.T., L.P., C.Z., G.P.P., G.T.), National & Kapodistrian University of Athens, "Attikon" University Hospital, Greece
| | - Konark Malhotra
- Department of Neurology, Allegheny Health Network, Pittsburgh, PA (K.M.)
| | - Christina Zompola
- Second Department of Neurology (A.T., L.P., C.Z., G.P.P., G.T.), National & Kapodistrian University of Athens, "Attikon" University Hospital, Greece
| | - Aristeidis H Katsanos
- Division of Neurology, McMaster University/Population Health Research Institute, Hamilton, Canada (A.H.K., A.S.)
| | - Ashkan Shoamanesh
- Division of Neurology, McMaster University/Population Health Research Institute, Hamilton, Canada (A.H.K., A.S.)
| | - Efstathios Boviatsis
- Department of Neurosurgery (E.B.), National & Kapodistrian University of Athens, "Attikon" University Hospital, Greece
| | - Efthimios Dardiotis
- Neurology Department, University Hospital of Larissa, University of Thessaly, Greece (E.D.)
| | - Martha Spilioti
- First Department of Neurology, AHEPA General Hospital, Aristotle University of Thessaloniki, Greece (M.S.)
| | - Simona Sacco
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, via Vetoio, Italy (S.S.)
| | - David J Werring
- Stroke Research Centre, UCL Queen Square Institute of Neurology, London, United Kingdom (D.J.W.)
| | - Charlotte Cordonnier
- University Lille, Inserm, CHU Lille, U1172, LilNCog, Lille Neuroscience and Cognition, France (C.C.)
| | - Andrei V Alexandrov
- Department of Neurology, University of Tennessee Health Science Center, Memphis (A.V.A., G.T.)
| | - George P Paraskevas
- Second Department of Neurology (A.T., L.P., C.Z., G.P.P., G.T.), National & Kapodistrian University of Athens, "Attikon" University Hospital, Greece
| | - Georgios Tsivgoulis
- Second Department of Neurology (A.T., L.P., C.Z., G.P.P., G.T.), National & Kapodistrian University of Athens, "Attikon" University Hospital, Greece.,Department of Neurology, University of Tennessee Health Science Center, Memphis (A.V.A., G.T.)
| |
Collapse
|
66
|
Reisz Z, Troakes C, Sztriha LK, Bodi I. Fatal thrombolysis-related intracerebral haemorrhage associated with amyloid-β-related angiitis in a middle-aged patient - case report and literature review. BMC Neurol 2022; 22:500. [PMID: 36564732 PMCID: PMC9783436 DOI: 10.1186/s12883-022-03029-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Amyloid-β-related angiitis (ABRA) is a rare complication of cerebral amyloid angiopathy, characterized by amyloid-β deposition in the leptomeningeal and cortical vessels with associated angiodestructive granulomatous inflammation. The clinical presentation is variable, including subacute cognitive decline, behavioural changes, headaches, seizures and focal neurological deficits, which may mimic other conditions. Here, we present a case with fatal thrombolysis-related haemorrhage associated with ABRA in a middle-aged patient. CASE PRESENTATION A 55-year-old man was admitted to hospital with sudden onset left-sided cheek, arm and hand sensory loss, blurred vision, and worsening headache, with a National Institutes of Health Stroke Scale (NIHSS) score of 3. An acute CT head scan showed no contraindications, and therefore the decision was made to give intravenous thrombolysis. Post-thrombolysis, he showed rapid deterioration with visual disturbances, headache and confusion, and a repeat CT head scan confirmed several areas of intracerebral haemorrhage. No benefit from surgical intervention was expected, and the patient died four days after the first presentation. Neuropathological examination found acute ischemic infarcts of three to five days duration in the basal ganglia, insular cortex and occipital lobe, correlating with the initial clinical symptoms. There were also extensive recent intracerebral haemorrhages most likely secondary to thrombolysis. Furthermore, the histological examination revealed severe cerebral amyloid angiopathy associated with granulomatous inflammatory reaction, consistent with ABRA. CONCLUSIONS Presentation of ABRA in a middle-aged patient highlighted the difficulties in recognition and management of this rare condition. There is emerging evidence that patients with CAA may have increased risk of fatal intracerebral haemorrhages following thrombolysis. This may be further increased by a coexisting CAA-related inflammatory vasculopathy which is potentially treatable with steroid therapy if early diagnosis is made.
Collapse
Affiliation(s)
- Zita Reisz
- grid.429705.d0000 0004 0489 4320Department of Clinical Neuropathology, King’s College Hospital NHS Foundation Trust, Denmark Hill, London, UK
| | - Claire Troakes
- grid.13097.3c0000 0001 2322 6764London Neurodegenerative Diseases Brain Bank, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| | - Laszlo K. Sztriha
- grid.429705.d0000 0004 0489 4320Department of Neurology, King’s College Hospital NHS Foundation Trust, Denmark Hill, London, UK
| | - Istvan Bodi
- grid.429705.d0000 0004 0489 4320Department of Clinical Neuropathology, King’s College Hospital NHS Foundation Trust, Denmark Hill, London, UK ,grid.13097.3c0000 0001 2322 6764London Neurodegenerative Diseases Brain Bank, Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, London, UK
| |
Collapse
|
67
|
Xia C, Lv Y. Cerebral amyloid angiopathy-related inflammation with posterior reversible encephalopathy syndrome-like presentation: a case report. BMC Neurol 2022; 22:449. [PMID: 36463107 PMCID: PMC9719169 DOI: 10.1186/s12883-022-02979-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 11/10/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Cerebral amyloid angiopathy-related inflammation (CAA-RI), which presents with acute or subacute cognitive or functional decline, focal or multifocal neurologic deficits, new onset of seizures, or a combination of seizures and neurologic deficits, shares clinical and radiologic similarities with posterior reversible encephalopathy syndrome (PRES). Differential diagnosis is critical because the treatment principle for these 2 conditions differs greatly. Here, we present a case of PRES-like CAA-RI and the strategy used to discriminate between the 2 conditions. CASE PRESENTATION A patient with probable CAA-RI was first thought to suffer from PRES. Initial high-dose methylprednisolone therapy caused rapid improvement of the neurologic symptoms but abrupt discontinuation of corticosteroids resulted in clinical relapse and deterioration. Subsequent reinitiation of high-dose methylprednisolone followed by tapering off of oral prednisone led to clinical and radiologic recovery at the 3-month follow-up. CONCLUSIONS We suggest that in cases where it is difficult to distinguish between CAA-RI and PRES solely based on magnetic resonance imaging, a good response to corticosteroids and an apolipoprotein E (ApoE) ε4/ε4 genotype are critical for establishing a diagnosis of CAA-RI. If there is clinical deterioration, sudden withdrawal of high-dose corticosteroid during the active phase of CAA-RI should be avoided.
Collapse
Affiliation(s)
- Cheng Xia
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang, China
| | - Yan Lv
- Department of Neurology, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
68
|
Klein G, Scelsi MA, Barakos J, Fiebach JB, Bracoud L, Suhy J, Delmar P, Lyons M, Wojtowicz J, Bullain S, Barkhof F, Purcell D. Comparing ARIA-E severity scales and effects of treatment management thresholds. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2022; 14:e12376. [PMID: 36474747 PMCID: PMC9716634 DOI: 10.1002/dad2.12376] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 10/14/2022] [Accepted: 10/19/2022] [Indexed: 11/25/2023]
Abstract
Introduction Amyloid-related imaging abnormalities-edema (ARIA-E) is associated with anti-amyloid beta monoclonal antibody treatment. ARIA-E severity may be assessed using the Barkhof Grand Total Scale (BGTS) or the 3- or 5-point Severity Scales of ARIA-E (SSAE-3/SSAE-5). We assessed inter- and intra-reader correlations between SSAE-3/5 and BGTS. Methods Magnetic resonance imaging scans were collected from 75 participants in the SCarlet RoAD and Marguerite RoAD studies. Three neuroradiologists reviewed scans at baseline and at follow-up. Concordance in dichotomized ARIA-E ratings was assessed for a range of BGTS thresholds. Results SSAE-3/5 scores correlated with BGTS scores, with high inter-reader intraclass correlation coefficients across all scales. There was high agreement in dichotomized ratings for SSAE-3 > 1 versus BGTS > 3 for all readers (accuracy 0.85-0.93) and between pairs of readers. Discussion SSAE-3/5 showed high degrees of correlation with BGTS, potentially allowing seamless transition from the BGTS to SSAE-3/5 for ARIA-E management.
Collapse
Affiliation(s)
| | | | - Jerome Barakos
- California Pacific Medical CenterSan FranciscoCaliforniaUSA
| | - Jochen B. Fiebach
- Center for Stroke Research BerlinCharité Universitätsmedizin BerlinBerlinGermany
| | - Luc Bracoud
- ClarioInc. (formerly Bioclinica, Inc.)LyonFrance
| | - Joyce Suhy
- ClarioInc. (formerly Bioclinica, Inc.)San MateoCaliforniaUSA
| | | | | | | | | | - Frederik Barkhof
- Department of Radiology & Nuclear MedicineAmsterdam UMCVrije UniversiteitAmsterdamNetherlands
- Queen Square Institute of Neurology and Centre for Medical Image ComputingUniversity College LondonLondonUK
| | - Derk Purcell
- California Pacific Medical CenterSan FranciscoCaliforniaUSA
| |
Collapse
|
69
|
Nehme A, Boulanger M, Aouba A, Pagnoux C, Zuber M, Touzé E, de Boysson H. Diagnostic and therapeutic approach to adult central nervous system vasculitis. Rev Neurol (Paris) 2022; 178:1041-1054. [PMID: 36156251 DOI: 10.1016/j.neurol.2022.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 05/16/2022] [Indexed: 12/14/2022]
Abstract
The clinical manifestations of central nervous system (CNS) vasculitis are highly variable. In the absence of a positive CNS biopsy, CNS vasculitis is particularly suspected when markers of both vascular disease and inflammation are present. To facilitate the clinical and therapeutic approach to this rare condition, CNS vasculitis can be classified according to the size of the involved vessels. Vascular imaging is used to identify medium vessel disease. Small vessel disease can only be diagnosed with a CNS biopsy. Medium vessel vasculitis usually presents with focal neurological signs, while small vessel vasculitis more often leads to cognitive deficits, altered level of consciousness and seizures. Markers of CNS inflammation include cerebrospinal fluid pleocytosis or elevated protein levels, and vessel wall, parenchymal or leptomeningeal enhancement. The broad range of differential diagnoses of CNS vasculitis can be narrowed based on the disease subtype. Common mimickers of medium vessel vasculitis include intracranial atherosclerosis and reversible cerebral vasoconstriction syndrome. The diagnostic workup aims to answer two questions: is the neurological presentation secondary to a vasculitic process, and if so, is the vasculitis primary (i.e., primary angiitis of the CNS) or secondary (e.g., to a systemic vasculitis, connective tissue disorder, infection, malignancy or drug use)? In primary angiitis of the CNS, glucocorticoids and cyclophosphamide are most often used for induction therapy, but rituximab may be an alternative. Based on the available evidence, all patients should receive maintenance immunosuppression. A multidisciplinary approach is necessary to ensure an accurate and timely diagnosis and to improve outcomes for patients with this potentially devastating condition.
Collapse
Affiliation(s)
- A Nehme
- Normandie University, Caen, France; Department of Neurology, Caen University Hospital, Caen, France; Inserm UMR-S U1237 PhIND/BB@C, Caen, France.
| | - M Boulanger
- Normandie University, Caen, France; Department of Neurology, Caen University Hospital, Caen, France; Inserm UMR-S U1237 PhIND/BB@C, Caen, France
| | - A Aouba
- Normandie University, Caen, France; Department of Internal Medicine, Caen University Hospital, Caen, France
| | - C Pagnoux
- Vasculitis clinic, Division of Rheumatology, Mount Sinai Hospital, University of Toronto, Toronto, Canada
| | - M Zuber
- Department of Neurology, Saint-Joseph Hospital, Paris, France; Université Paris Cité, Paris, France
| | - E Touzé
- Normandie University, Caen, France; Department of Neurology, Caen University Hospital, Caen, France; Inserm UMR-S U1237 PhIND/BB@C, Caen, France
| | - H de Boysson
- Normandie University, Caen, France; Department of Internal Medicine, Caen University Hospital, Caen, France
| |
Collapse
|
70
|
Kelly MA, Singh J, Balabanov A, Wadina A, Dasovic B. Challenging Cases in Cerebrovascular Disease. Semin Neurol 2022; 42:758-766. [PMID: 36417992 DOI: 10.1055/a-1985-7000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Disorders involving the vascular system of the brain are numerous and sundry. Atherosclerotic thromboembolism of large vessels and lacunar infarctions of small vessel disease are well known. Brain infarction due to cardioembolism is common as well, and even more so when diligently sought. Rupture of intracranial blood vessels results in subarachnoid and intraparenchymal hemorrhage. We present four cases of stroke of uncommon cause and remind clinicians to be open minded to the many possible causes of stroke, in particular because early recognition and treatment is often critical. Case 1 discusses a patient with inflammatory cerebral amyloid angiopathy. The presentation, ability to recur, and current treatment considerations are reviewed. Case 2 discusses microangiopathic thrombotic angiopathy. Diagnosis and treatment are considered. An association with interferon therapy and the evolving terminology of this and related conditions are discussed. Case 3 discusses intracranial hemorrhage secondary to acute promyelocytic leukemia. Patients with acute leukemias require aggressive management of their coagulopathy, thrombocytopenia, and the disease itself. Finally, Case 4 discusses ischemic stroke due to a paradoxical embolism in the setting of a patent foramen ovale (PFO). Both medical and surgical management of a PFO for stroke prevention are considered.
Collapse
Affiliation(s)
- Michael A Kelly
- Department of Neurology, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois
| | - Jasmine Singh
- Department of Neurology, University of California San Diego Ringgold Standard Institution, La Jolla, California
| | - Alexandra Balabanov
- Department of Neurology, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois
| | - Adam Wadina
- Department of Neurology, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois
| | - Braden Dasovic
- Department of Neurology, Loyola University Chicago, Stritch School of Medicine, Maywood, Illinois
| |
Collapse
|
71
|
Cerebral Superficial Siderosis. Clin Neuroradiol 2022; 33:293-306. [DOI: 10.1007/s00062-022-01231-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 10/11/2022] [Indexed: 11/29/2022]
Abstract
AbstractSuperficial siderosis (SS) of the central nervous system constitutes linear hemosiderin deposits in the leptomeninges and the superficial layers of the cerebrum and the spinal cord. Infratentorial (i) SS is likely due to recurrent or continuous slight bleeding into the subarachnoid space. It is assumed that spinal dural pathologies often resulting in cerebrospinal fluid (CSF) leakage is the most important etiological group which causes iSS and detailed neuroradiological assessment of the spinal compartment is necessary. Further etiologies are neurosurgical interventions, trauma and arteriovenous malformations. Typical neurological manifestations of this classical type of iSS are slowly progressive sensorineural hearing impairment and cerebellar symptoms, such as ataxia, kinetic tremor, nystagmus and dysarthria. Beside iSS, a different type of SS restricted to the supratentorial compartment can be differentiated, i.e. cortical (c) SS, especially in older people often due to cerebral amyloid angiopathy (CAA). Clinical presentation of cSS includes transient focal neurological episodes or “amyloid spells”. In addition, spontaneous and amyloid beta immunotherapy-associated CAA-related inflammation may cause cSS, which is included in the hemorrhagic subgroup of amyloid-related imaging abnormalities (ARIA). Because a definitive diagnosis requires a brain biopsy, knowledge of neuroimaging features and clinical findings in CAA-related inflammation is essential. This review provides neuroradiological hallmarks of the two groups of SS and give an overview of neurological symptoms and differential diagnostic considerations.
Collapse
|
72
|
Theodorou A, Palaiodimou L, Safouris A, Kargiotis O, Psychogios K, Kotsali-Peteinelli V, Foska A, Zouvelou V, Tzavellas E, Tzanetakos D, Zompola C, Tzartos JS, Voumvourakis K, Paraskevas GP, Tsivgoulis G. Cerebral Amyloid Angiopathy-Related Inflammation: A Single-Center Experience and a Literature Review. J Clin Med 2022; 11:6731. [PMID: 36431207 PMCID: PMC9692654 DOI: 10.3390/jcm11226731] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
Background: Limited data exist regarding the prevalence of clinical, neuroimaging, and genetic markers among patients diagnosed with Cerebral Amyloid Angiopathy−related inflammation (CAA-ri). We sought to determine these characteristics in patients diagnosed in our center and to summarize available literature published either as single-case reports or small case series (<5 patients). Methods: We reported our single-center experience of patients diagnosed with CAA-ri according to international criteria during a seven-year period (2015−2022), and we abstracted data from 90 previously published cases. Results: Seven patients (43% women, mean age 70 ± 13 years) were diagnosed with CAA-ri in our center. The most common symptom at presentation was focal neurological dysfunction (71%), and the most prevalent radiological finding was the presence of T2/FLAIR white matter hyperintensities (100%). All patients were treated with corticosteroids and had a favorable functional outcome. Among 90 previously published CAA-ri cases (51% women, mean age 70 ± 9 years), focal neurological dysfunction was the most common symptom (76%), followed by a cognitive decline (46%) and headache (34%). The most prevalent neuroimaging findings were cerebral microbleeds (85%), asymmetric T2/FLAIR white matter hyperintensities (81%), and gadolinium-enhancing T1-lesions (37%). Genetic testing for the Apolipoprotein-E gene was available in 27 cases; 59% carried the APOE ε4/ε4 genotype. The majority of the published CAA-ri cases (78%) received corticosteroid monotherapy, while 17 patients (19%) were treated with additional immunosuppressive treatment. Favorable functional outcome following treatment was documented in 70% of patients. Conclusion: Improving the vigilance of clinicians regarding the early recognition and accurate diagnosis of CAA-ri is crucial for swift therapy initiation, which may result in improved functional outcomes.
Collapse
Affiliation(s)
- Aikaterini Theodorou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Lina Palaiodimou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Apostolos Safouris
- Stroke Unit, Metropolitan Hospital, Ethnarhou Makariou 9, N. Faliro, 18547 Piraeus, Greece
| | - Odysseas Kargiotis
- Stroke Unit, Metropolitan Hospital, Ethnarhou Makariou 9, N. Faliro, 18547 Piraeus, Greece
| | - Klearchos Psychogios
- Stroke Unit, Metropolitan Hospital, Ethnarhou Makariou 9, N. Faliro, 18547 Piraeus, Greece
| | - Vasiliki Kotsali-Peteinelli
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Aikaterini Foska
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Vasiliki Zouvelou
- First Department of Neurology, “Aiginition” Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Elias Tzavellas
- First Department of Psychiatry, “Aiginition” Hospital, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Dimitrios Tzanetakos
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Christina Zompola
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - John S. Tzartos
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Konstantinos Voumvourakis
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Georgios P. Paraskevas
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - Georgios Tsivgoulis
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
73
|
Imaging of cerebral amyloid angiopathy–related inflammation (CAA-ri). Neurol Sci 2022; 43:6151-6153. [DOI: 10.1007/s10072-022-06189-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 05/31/2022] [Indexed: 10/18/2022]
|
74
|
Piazza F, Caminiti SP, Zedde M, Presotto L, DiFrancesco JC, Pascarella R, Giossi A, Sessa M, Poli L, Basso G, Perani D. Association of Microglial Activation With Spontaneous ARIA-E and CSF Levels of Anti-Aβ Autoantibodies. Neurology 2022; 99:e1265-e1277. [PMID: 35940900 PMCID: PMC9576297 DOI: 10.1212/wnl.0000000000200892] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Accepted: 05/13/2022] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Amyloid-related imaging abnormalities suggestive of vasogenic edema or sulcal effusion (ARIA-E) are the most common adverse events complicating Alzheimer disease (AD) immunotherapy with anti-β-amyloid (Aβ) monoclonal antibodies. ARIA-E can also occur spontaneously in cerebral amyloid angiopathy-related inflammation (CAA-ri), a rare autoimmune encephalopathy associated with increased CSF levels of anti-Aβ autoantibodies. Although the pathophysiologic mechanisms of ARIA-E remain to be fully elucidated, experimental evidence from ex vivo studies suggests that gantenerumab and aducanumab enable microglial activation. However, the in vivo evidence for a direct association between neuroinflammation and ARIA-E in patients with high CSF anti-Aβ (auto)antibody levels has never been demonstrated. METHODS The spatial distribution and temporal variations of microglial activation associated with levels of anti-Aβ autoantibodies at (sub)acute presentation of ARIA-E and after corticosteroid therapy were evaluated in a longitudinal case series of patients with CAA-ri, the spontaneous variant of the iatrogenic ARIA-E reported in Aβ-lowering immunotherapy with monoclonal antibodies. Multimodal and multiparametric MRI was used for CAA and ARIA-E severity quantification, according to validated scoring system; CSF testing for anti-Aβ autoantibodies and AD biomarkers; 11C-PK11195 PET for activated microglia. RESULTS At (sub)acute presentation, we found focal peaks of microglial activation having a greater spatial colocalization with ARIA-E compared with chronic age-related white matter change imaging abnormalities. The severity of ARIA-E and the magnitude of the associated microglial activation were greater in patients having AD and severe CAA concomitant disease compared with patients having CAA only. CSF anti-Aβ autoantibodies at presentation were high in all patients and markedly decreased at posttreatment follow-up, in parallel with clinical resolution of acute symptoms, reduced ARIA-E severity, and reduced microglial activation. DISCUSSION Our findings extend the current notion of ARIA-E by providing the first in vivo 11C-PK11195 PET evidence for an association between microglial activation and the magnitude and severity of ARIA-E in patients with increased CSF concentration of anti-Aβ autoantibodies and comorbid AD and CAA disease. Our results highlight CSF testing for anti-Aβ autoantibodies as a promising diagnostic, prognostic, and therapy response biomarker to help guide future treatment and management decisions in real clinical practice and clinical trials.
Collapse
Affiliation(s)
- Fabrizio Piazza
- From the CAA and AD Translational Research and Biomarkers Laboratory (F.P.), School of Medicine and Surgery (J.C.D., G.B.), iCAβ International Network (F.P., M.Z., J.C.D., A.G., M.S., Loris Poli, G.B.), and SINdem CAA Study Group (F.P., M.Z., D.P.), University of Milano-Bicocca, Monza; Vita-Salute San Raffaele University (S.P.C., D.P.), Milan; IRCCS San Raffaele Scientific Institute (S.P.C., Luca Presotto, D.P.), Milan; Neurology Unit (M.Z.), Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neuroradiology Unit (R.P.), Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neurology Unit (A.G.), Azienda Socio-Sanitaria Territoriale di Cremona; Neurology Unit (M.S.), Ospedale ASST Papa Giovanni XXIII, Bergamo; and Neurology Unit (Loris Poli), ASST Spedali Civili, Brescia, Italy.
| | - Silvia Paola Caminiti
- From the CAA and AD Translational Research and Biomarkers Laboratory (F.P.), School of Medicine and Surgery (J.C.D., G.B.), iCAβ International Network (F.P., M.Z., J.C.D., A.G., M.S., Loris Poli, G.B.), and SINdem CAA Study Group (F.P., M.Z., D.P.), University of Milano-Bicocca, Monza; Vita-Salute San Raffaele University (S.P.C., D.P.), Milan; IRCCS San Raffaele Scientific Institute (S.P.C., Luca Presotto, D.P.), Milan; Neurology Unit (M.Z.), Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neuroradiology Unit (R.P.), Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neurology Unit (A.G.), Azienda Socio-Sanitaria Territoriale di Cremona; Neurology Unit (M.S.), Ospedale ASST Papa Giovanni XXIII, Bergamo; and Neurology Unit (Loris Poli), ASST Spedali Civili, Brescia, Italy
| | - Marialuisa Zedde
- From the CAA and AD Translational Research and Biomarkers Laboratory (F.P.), School of Medicine and Surgery (J.C.D., G.B.), iCAβ International Network (F.P., M.Z., J.C.D., A.G., M.S., Loris Poli, G.B.), and SINdem CAA Study Group (F.P., M.Z., D.P.), University of Milano-Bicocca, Monza; Vita-Salute San Raffaele University (S.P.C., D.P.), Milan; IRCCS San Raffaele Scientific Institute (S.P.C., Luca Presotto, D.P.), Milan; Neurology Unit (M.Z.), Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neuroradiology Unit (R.P.), Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neurology Unit (A.G.), Azienda Socio-Sanitaria Territoriale di Cremona; Neurology Unit (M.S.), Ospedale ASST Papa Giovanni XXIII, Bergamo; and Neurology Unit (Loris Poli), ASST Spedali Civili, Brescia, Italy
| | - Luca Presotto
- From the CAA and AD Translational Research and Biomarkers Laboratory (F.P.), School of Medicine and Surgery (J.C.D., G.B.), iCAβ International Network (F.P., M.Z., J.C.D., A.G., M.S., Loris Poli, G.B.), and SINdem CAA Study Group (F.P., M.Z., D.P.), University of Milano-Bicocca, Monza; Vita-Salute San Raffaele University (S.P.C., D.P.), Milan; IRCCS San Raffaele Scientific Institute (S.P.C., Luca Presotto, D.P.), Milan; Neurology Unit (M.Z.), Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neuroradiology Unit (R.P.), Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neurology Unit (A.G.), Azienda Socio-Sanitaria Territoriale di Cremona; Neurology Unit (M.S.), Ospedale ASST Papa Giovanni XXIII, Bergamo; and Neurology Unit (Loris Poli), ASST Spedali Civili, Brescia, Italy
| | - Jacopo C DiFrancesco
- From the CAA and AD Translational Research and Biomarkers Laboratory (F.P.), School of Medicine and Surgery (J.C.D., G.B.), iCAβ International Network (F.P., M.Z., J.C.D., A.G., M.S., Loris Poli, G.B.), and SINdem CAA Study Group (F.P., M.Z., D.P.), University of Milano-Bicocca, Monza; Vita-Salute San Raffaele University (S.P.C., D.P.), Milan; IRCCS San Raffaele Scientific Institute (S.P.C., Luca Presotto, D.P.), Milan; Neurology Unit (M.Z.), Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neuroradiology Unit (R.P.), Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neurology Unit (A.G.), Azienda Socio-Sanitaria Territoriale di Cremona; Neurology Unit (M.S.), Ospedale ASST Papa Giovanni XXIII, Bergamo; and Neurology Unit (Loris Poli), ASST Spedali Civili, Brescia, Italy
| | - Rosario Pascarella
- From the CAA and AD Translational Research and Biomarkers Laboratory (F.P.), School of Medicine and Surgery (J.C.D., G.B.), iCAβ International Network (F.P., M.Z., J.C.D., A.G., M.S., Loris Poli, G.B.), and SINdem CAA Study Group (F.P., M.Z., D.P.), University of Milano-Bicocca, Monza; Vita-Salute San Raffaele University (S.P.C., D.P.), Milan; IRCCS San Raffaele Scientific Institute (S.P.C., Luca Presotto, D.P.), Milan; Neurology Unit (M.Z.), Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neuroradiology Unit (R.P.), Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neurology Unit (A.G.), Azienda Socio-Sanitaria Territoriale di Cremona; Neurology Unit (M.S.), Ospedale ASST Papa Giovanni XXIII, Bergamo; and Neurology Unit (Loris Poli), ASST Spedali Civili, Brescia, Italy
| | - Alessia Giossi
- From the CAA and AD Translational Research and Biomarkers Laboratory (F.P.), School of Medicine and Surgery (J.C.D., G.B.), iCAβ International Network (F.P., M.Z., J.C.D., A.G., M.S., Loris Poli, G.B.), and SINdem CAA Study Group (F.P., M.Z., D.P.), University of Milano-Bicocca, Monza; Vita-Salute San Raffaele University (S.P.C., D.P.), Milan; IRCCS San Raffaele Scientific Institute (S.P.C., Luca Presotto, D.P.), Milan; Neurology Unit (M.Z.), Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neuroradiology Unit (R.P.), Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neurology Unit (A.G.), Azienda Socio-Sanitaria Territoriale di Cremona; Neurology Unit (M.S.), Ospedale ASST Papa Giovanni XXIII, Bergamo; and Neurology Unit (Loris Poli), ASST Spedali Civili, Brescia, Italy
| | - Maria Sessa
- From the CAA and AD Translational Research and Biomarkers Laboratory (F.P.), School of Medicine and Surgery (J.C.D., G.B.), iCAβ International Network (F.P., M.Z., J.C.D., A.G., M.S., Loris Poli, G.B.), and SINdem CAA Study Group (F.P., M.Z., D.P.), University of Milano-Bicocca, Monza; Vita-Salute San Raffaele University (S.P.C., D.P.), Milan; IRCCS San Raffaele Scientific Institute (S.P.C., Luca Presotto, D.P.), Milan; Neurology Unit (M.Z.), Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neuroradiology Unit (R.P.), Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neurology Unit (A.G.), Azienda Socio-Sanitaria Territoriale di Cremona; Neurology Unit (M.S.), Ospedale ASST Papa Giovanni XXIII, Bergamo; and Neurology Unit (Loris Poli), ASST Spedali Civili, Brescia, Italy
| | - Loris Poli
- From the CAA and AD Translational Research and Biomarkers Laboratory (F.P.), School of Medicine and Surgery (J.C.D., G.B.), iCAβ International Network (F.P., M.Z., J.C.D., A.G., M.S., Loris Poli, G.B.), and SINdem CAA Study Group (F.P., M.Z., D.P.), University of Milano-Bicocca, Monza; Vita-Salute San Raffaele University (S.P.C., D.P.), Milan; IRCCS San Raffaele Scientific Institute (S.P.C., Luca Presotto, D.P.), Milan; Neurology Unit (M.Z.), Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neuroradiology Unit (R.P.), Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neurology Unit (A.G.), Azienda Socio-Sanitaria Territoriale di Cremona; Neurology Unit (M.S.), Ospedale ASST Papa Giovanni XXIII, Bergamo; and Neurology Unit (Loris Poli), ASST Spedali Civili, Brescia, Italy
| | - Gianpaolo Basso
- From the CAA and AD Translational Research and Biomarkers Laboratory (F.P.), School of Medicine and Surgery (J.C.D., G.B.), iCAβ International Network (F.P., M.Z., J.C.D., A.G., M.S., Loris Poli, G.B.), and SINdem CAA Study Group (F.P., M.Z., D.P.), University of Milano-Bicocca, Monza; Vita-Salute San Raffaele University (S.P.C., D.P.), Milan; IRCCS San Raffaele Scientific Institute (S.P.C., Luca Presotto, D.P.), Milan; Neurology Unit (M.Z.), Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neuroradiology Unit (R.P.), Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neurology Unit (A.G.), Azienda Socio-Sanitaria Territoriale di Cremona; Neurology Unit (M.S.), Ospedale ASST Papa Giovanni XXIII, Bergamo; and Neurology Unit (Loris Poli), ASST Spedali Civili, Brescia, Italy
| | - Daniela Perani
- From the CAA and AD Translational Research and Biomarkers Laboratory (F.P.), School of Medicine and Surgery (J.C.D., G.B.), iCAβ International Network (F.P., M.Z., J.C.D., A.G., M.S., Loris Poli, G.B.), and SINdem CAA Study Group (F.P., M.Z., D.P.), University of Milano-Bicocca, Monza; Vita-Salute San Raffaele University (S.P.C., D.P.), Milan; IRCCS San Raffaele Scientific Institute (S.P.C., Luca Presotto, D.P.), Milan; Neurology Unit (M.Z.), Stroke Unit, Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neuroradiology Unit (R.P.), Azienda Unità Sanitaria Locale-IRCCS di Reggio Emilia; Neurology Unit (A.G.), Azienda Socio-Sanitaria Territoriale di Cremona; Neurology Unit (M.S.), Ospedale ASST Papa Giovanni XXIII, Bergamo; and Neurology Unit (Loris Poli), ASST Spedali Civili, Brescia, Italy
| |
Collapse
|
75
|
Cogswell PM, Barakos JA, Barkhof F, Benzinger TS, Jack CR, Poussaint TY, Raji CA, Ramanan VK, Whitlow CT. Amyloid-Related Imaging Abnormalities with Emerging Alzheimer Disease Therapeutics: Detection and Reporting Recommendations for Clinical Practice. AJNR Am J Neuroradiol 2022; 43:E19-E35. [PMID: 35953274 PMCID: PMC9451628 DOI: 10.3174/ajnr.a7586] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Monoclonal antibodies are emerging disease-modifying therapies for Alzheimer disease that require brain MR imaging for eligibility assessment as well as for monitoring for amyloid-related imaging abnormalities. Amyloid-related imaging abnormalities result from treatment-related loss of vascular integrity and may occur in 2 forms. Amyloid-related imaging abnormalities with edema or effusion are transient, treatment-induced edema or sulcal effusion, identified on T2-FLAIR. Amyloid-related imaging abnormalities with hemorrhage are treatment-induced microhemorrhages or superficial siderosis identified on T2* gradient recalled-echo. As monoclonal antibodies become more widely available, treatment screening and monitoring brain MR imaging examinations may greatly increase neuroradiology practice volumes. Radiologists must become familiar with the imaging appearance of amyloid-related imaging abnormalities, how to select an appropriate imaging protocol, and report findings in clinical practice. On the basis of clinical trial literature and expert experience from clinical trial imaging, we summarize imaging findings of amyloid-related imaging abnormalities, describe potential interpretation pitfalls, and provide recommendations for a standardized imaging protocol and an amyloid-related imaging abnormalities reporting template. Standardized imaging and reporting of these findings are important because an amyloid-related imaging abnormalities severity score, derived from the imaging findings, is used along with clinical status to determine patient management and eligibility for continued monoclonal antibody dosing.
Collapse
Affiliation(s)
- P M Cogswell
- From the Departments of Radiology (P.M.C., C.R.J.)
| | - J A Barakos
- Department of Radiology (J.A.B.), California Pacific Medical Center, San Francisco, California
| | - F Barkhof
- Departments of Radiology (F.B.)
- Nuclear Medicine (F.B.), VU University Medical Center, Amsterdam, the Netherlands
- Queen Square Institute of Neurology and Centre for Medical Image Computing (F.B.), University College London, UK
| | - T S Benzinger
- Departments of Radiology (T.S.B., C.A.R.)
- Neurosurgery (T.S.B.)
| | - C R Jack
- From the Departments of Radiology (P.M.C., C.R.J.)
| | - T Y Poussaint
- Department of Radiology (T.Y.P.), Boston Children's Hospital, Boston, Massachusetts
| | - C A Raji
- Departments of Radiology (T.S.B., C.A.R.)
- Neurology (C.A.R.),Washington University School of Medicine, St. Louis, Missouri
| | - V K Ramanan
- Neurology (V.K.R.), Mayo Clinic, Rochester, Minnesota
| | - C T Whitlow
- Departments of Radiology (C.T.W.)
- Biomedical Engineering (C.T.W.), Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
76
|
Diagnosis, treatment, and follow-up of patients with cerebral amyloid angiopathy-related inflammation. Neurol Sci 2022; 43:6381-6387. [PMID: 35930182 PMCID: PMC9616779 DOI: 10.1007/s10072-022-06299-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 07/20/2022] [Indexed: 11/09/2022]
Abstract
Purpose Cerebral amyloid angiopathy-related inflammation (CAA-ri) is a rare potentially reversible encephalopathy associated with an autoimmune process against proteins deposited in the walls of cortical and leptomeningeal brain vessels. Definite diagnosis requires histopathological features of vascular inflammation and amyloid deposition from brain biopsy. Clinical-neuroradiological criteria have been recently introduced and validated to reduce the need for biopsy. The purpose of this paper is to report a historical retrospective review of clinical-neuroradiological follow-up of two patients with probable CAA-ri and five patients with a reasonably probable suspect of CAA-ri (4 females, 3 males, patient’s age at admission: 66–79 years) seen at our institution between 2007 and 2021, focusing on clinical and neuroradiological awareness to this entity and variable response to immunotherapy. Materials and methods Clinical features at presentation included subacute to acute confusion (6/7), seizures (4/7), cognitive impairment (5/7), and focal neurological signs (3/7). Neuroradiology included braincomputed tomography followed by magnetic resonance imaging. Infectious diseases and autoimmune workups were then performed. Results CSF analysis was performed in two patients. Cerebral angiography was performed in two patients, to rule out vascular malformations. Hemorrhagic posterior reversible encephalopathy syndrome has been suspected in two patients. Four patients underwent immunotherapy with corticosteroids followed by reduction of brain dysfunctions. Three patients did not undergo immunotherapy but underwent clinical and/or neuroradiological remission. Conclusions Patients with CAA-ri present a rare steroid-responsive acute to subacute brain dysfunction. Thus, it has to be known and recognized both clinically and neuroradiologically. Spontaneous clinical and/or neuroradiological improvement is possible in patients with mild symptoms.
Collapse
|
77
|
Grangeon L, Quesney G, Verdalle-Cazes M, Coulette S, Renard D, Wacongne A, Allou T, Olivier N, Boukriche Y, Blanchet-Fourcade G, Labauge P, Arquizan C, Canaple S, Godefroy O, Martinaud O, Verdure P, Quillard-Muraine M, Pariente J, Magnin E, Nicolas G, Charbonnier C, Maltête D, Formaglio M, Raposo N, Ayrignac X, Wallon D. Different clinical outcomes between cerebral amyloid angiopathy-related inflammation and non-inflammatory form. J Neurol 2022; 269:4972-4984. [PMID: 35752990 DOI: 10.1007/s00415-022-11145-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 10/17/2022]
Abstract
OBJECTIVE Cerebral amyloid angiopathy-related inflammation (CAA-ri) is a rare manifestation related to CAA, thought to be more severe. We aimed to compare the clinical and radiological outcomes of CAA-ri and non-inflammatory CAA. MATERIALS AND METHODS We retrospectively included all patients with CAA-ri from 13 French centers. We constituted a sex- and age-matched control cohort with non-inflammatory CAA and similar disease duration. Survival, autonomy and cognitive evolution were compared after logistic regression. Cerebral microbleeds (CMB), intracerebral hemorrhage, cortical superficial siderosis and hippocampal atrophy were analyzed as well as CSF biomarker profile and APOE genotype when available. Outcomes were compared using Kaplan-Meier curves and log-rank tests. RESULTS Data from 48 CAA-ri patients including 28 already reported and 20 new patients were analyzed. Over a mean of 3.1 years, 11 patients died (22.9%) and 18 (37.5%) relapsed. CAA-ri patients were more frequently institutionalized than non-inflammatory CAA patients (30% vs 8.3%, p < 0.001); mortality rates remained similar. MMSE and modified Rankin scale scores showed greater severity in CAA-ri at last follow-up. MRI showed a higher number of CMB at baseline and last follow-up in CAA-ri (p < 0.001 and p = 0.004, respectively). CSF showed lower baseline levels of Aß42 in CAA-ri than non-inflammatory CAA (373.3 pg/ml vs 490.8 pg/ml, p = 0.05). CAA-ri patients more likely carried at least one APOE ε4 allele (76% vs 37.5%, adjusted p = 0.05) particularly as homozygous status (56% vs 6.2%, p < 0.001). INTERPRETATION CAA-ri appears to be more severe than non-inflammatory CAA with a significant loss of autonomy and global higher amyloid burden, shown by more CMB and a distinct CSF profile. This burden may be partially promoted by ε4 allele.
Collapse
Affiliation(s)
- L Grangeon
- Department of Neurology, Rouen University Hospital, 76031, Rouen, France.
| | - G Quesney
- Department of Neurology, Rouen University Hospital, 76031, Rouen, France
| | - M Verdalle-Cazes
- Department of Radiology, Rouen University Hospital, Rouen, France
| | - S Coulette
- Department of Neurology, INM, Univ Montpellier, INSERM, Montpellier University Hospital, Montpellier, France
| | - D Renard
- Department of Neurology, Nimes University Hospital, Nimes, France
| | - A Wacongne
- Department of Neurology, Nimes University Hospital, Nimes, France
| | - T Allou
- Department of Neurology, Perpignan Hospital, Perpignan, France
| | - N Olivier
- Department of Neurology, Perpignan Hospital, Perpignan, France
| | - Y Boukriche
- Department of Neurology, Beziers Hospital, Beziers, France
| | | | - P Labauge
- Department of Neurology, INM, Univ Montpellier, INSERM, Montpellier University Hospital, Montpellier, France
| | - C Arquizan
- Department of Neurology, INM, Univ Montpellier, INSERM, Montpellier University Hospital, Montpellier, France
| | - S Canaple
- Department of Neurology and Functional Neuroscience, Lab (UR UPJV 4559), Amiens University Hospital and University of Picardy Jules Verne, Amiens, France
| | - O Godefroy
- Department of Neurology and Functional Neuroscience, Lab (UR UPJV 4559), Amiens University Hospital and University of Picardy Jules Verne, Amiens, France
| | - O Martinaud
- Department of Neurology, Caen University Hospital, Caen, France.,EPHE, INSERM, U1077, CHU de Caen, Neuropsychologie Et Imagerie de La Mémoire Humaine, Normandie Univ, UNICAEN, PSL Research University, Caen, France
| | - P Verdure
- Department of Neurology, Les Feugrais Hospital, Elbeuf, France
| | - M Quillard-Muraine
- Laboratoire de Biochimie, Rouen University Hospital and University of Rouen, Rouen, France
| | - J Pariente
- Neurology Department, Hôpital Pierre-Paul Riquet, Centre Hospitalier, Universitaire de Toulouse, Toulouse, France
| | - E Magnin
- Department of Neurology, Besancon Hospital, Besancon, France
| | - G Nicolas
- INSERM U1245, IRIB, Normandy University, CNR-MAJ, Rouen University Hospital, Rouen, France
| | - C Charbonnier
- INSERM U1245, IRIB, Normandy University, CNR-MAJ, Rouen University Hospital, Rouen, France
| | - D Maltête
- Department of Neurology, Rouen University Hospital, 76031, Rouen, France
| | - M Formaglio
- Department of Neurology, Lyon University Hospital, Lyon, France
| | - N Raposo
- Neurology Department, Hôpital Pierre-Paul Riquet, Centre Hospitalier, Universitaire de Toulouse, Toulouse, France
| | - X Ayrignac
- Department of Neurology, INM, Univ Montpellier, INSERM, Montpellier University Hospital, Montpellier, France
| | - D Wallon
- Department of Neurology, Rouen University Hospital, 76031, Rouen, France.,Department of Neurology, Besancon Hospital, Besancon, France
| |
Collapse
|
78
|
Situ M, Citalan-Madrid AF, Stamatovic SM, Keep RF, Andjelkovic AV. Transcriptomic Profile of Blood–Brain Barrier Remodeling in Cerebral Amyloid Angiopathy. Front Cell Neurosci 2022; 16:931247. [PMID: 35813502 PMCID: PMC9257207 DOI: 10.3389/fncel.2022.931247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 05/30/2022] [Indexed: 12/16/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) is a small vessel disease characterized by amyloid β (Aβ) peptide deposition within the walls of medium to small-caliber blood vessels, cerebral microhemorrhage, and blood–brain barrier (BBB) leakage. It is commonly associated with late-stage Alzheimer’s disease. BBB dysfunction is indicated as a pathological substrate for CAA progression with hyperpermeability, enhancing the extravasation of plasma components and inducing neuroinflammation, further worsening BBB injury and contributing to cognitive decline. Although significant effort has been made in defining the gene mutations and risk factors involved in microvascular alterations with vascular dementia and Alzheimer’s disease, the intra- and intercellular pathogenic mechanisms responsible for vascular hyperpermeability are still largely unknown. The present study aimed to elucidate the transcriptional profile of the cerebral microvessels (BBB) in a murine model with CAA vasculopathy to define potential causes and underlying mechanisms of BBB injury. A comprehensive RNA sequencing analysis was performed of CAA vasculopathy in Tg-SwDI mice at 6 and 18 months in comparison to age-matched wildtype controls to examine how age and amyloid accumulation impact the transcriptional signature of the BBB. Results indicate that Aβ has a critical role in triggering brain endothelial cell and BBB dysfunction in CAA vasculopathy, causing an intense proinflammatory response, impairing oxidative metabolism, altering the coagulation status of brain endothelial cells, and remodeling barrier properties. The proinflammatory response includes both adaptive and innate immunity, with pronounced induction of genes that regulate macrophage/microglial activation and chemokines/adhesion molecules that support T and B cell transmigration. Age has an important impact on the effects of Aβ, increasing the BBB injury in CAA vasculopathy. However, early inflammation, particularly microglia/macrophage activation and the mediators of B lymphocytes’ activities are underlying processes of BBB hyperpermeability and cerebral microbleeds in the early stage of CAA vasculopathy. These findings reveal a specific profile of the CAA-associated BBB injury that leads to a full progression of CAA.
Collapse
Affiliation(s)
- Muyu Situ
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
| | | | - Svetlana M. Stamatovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Richard F. Keep
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Anuska V. Andjelkovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- *Correspondence: Anuska V. Andjelkovic,
| |
Collapse
|
79
|
Dudley A, Sweeney K, Looby S, Farrell M, McGovern E. Teaching NeuroImage: Cerebral Amyloid Angiopathy Related Inflammation: An Interesting Evolution of Imaging Findings. Neurology 2022; 99:216-217. [PMID: 35654596 DOI: 10.1212/wnl.0000000000200833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/22/2022] [Indexed: 11/15/2022] Open
Affiliation(s)
- Alex Dudley
- )Department of Neurology, Beaumont Hospital, Dublin 9, Ireland
| | - Kieron Sweeney
- )National centre for Neurosurgery , Beaumont Hospital, Dublin 9, Ireland.,)Honorary senior clinical lecturer, Royal College of Surgeons in Ireland
| | - Seamus Looby
- )Department of Radiology, Beaumont Hospital, Dublin 9, Ireland
| | - Michael Farrell
- )Department of Neuropathology, Beaumont Hospital, Dublin 9, Ireland
| | - Eavan McGovern
- )Department of Neurology, Beaumont Hospital, Dublin 9, Ireland.,)Honorary senior clinical lecturer, Royal College of Surgeons in Ireland
| |
Collapse
|
80
|
Piazza F, Frölich L, Padovani A. Large Collaborative Registries and Real-world Data to Manage Amyloid-Related Imaging Abnormalities. JAMA Neurol 2022; 79:633-634. [PMID: 35467713 DOI: 10.1001/jamaneurol.2022.0731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Fabrizio Piazza
- iCAB International Network, CAA and AD Translational Research Biomarkers, School of Medicine, University of Milano-Bicocca, Monza, Italy
| | - Lutz Frölich
- Department of Geriatric Psychiatry, Central Institute of Mental Health, Medical Faculty Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Alessandro Padovani
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
81
|
DiFrancesco JC, Labate A, Romoli M, Chipi E, Salvadori N, Galimberti CA, Perani D, Ferrarese C, Costa C. Clinical and Instrumental Characterization of Patients With Late-Onset Epilepsy. Front Neurol 2022; 13:851897. [PMID: 35359649 PMCID: PMC8963711 DOI: 10.3389/fneur.2022.851897] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 02/04/2022] [Indexed: 11/13/2022] Open
Abstract
Epilepsy is classically considered a childhood disease. However, it represents the third most frequent neurological condition in the elderly, following stroke, and dementia. With the progressive aging of the general population, the number of patients with Late-Onset Epilepsy (LOE) is constantly growing, with important economic and social consequences, in particular for the more developed countries where the percentage of elderly people is higher. The most common causes of LOE are structural, mainly secondary to cerebrovascular or infectious diseases, brain tumors, trauma, and metabolic or toxic conditions. Moreover, there is a growing body of evidence linking LOE with neurodegenerative diseases, particularly Alzheimer's disease (AD). However, despite a thorough characterization, the causes of LOE remain unknown in a considerable portion of patients, thus termed as Late-Onset Epilepsy of Unknown origin (LOEU). In order to identify the possible causes of the disease, with an important impact in terms of treatment and prognosis, LOE patients should always undergo an exhaustive phenotypic characterization. In this work, we provide a detailed review of the main clinical and instrumental techniques for the adequate characterization of LOE patients in the clinical practice. This work aims to provide an easy and effective tool that supports routine activity of the clinicians facing LOE.
Collapse
Affiliation(s)
- Jacopo C. DiFrancesco
- Department of Neurology, ASST S. Gerardo Hospital, School of Medicine and Surgery and Milan Center for Neuroscience, University of Milano - Bicocca, Monza, Italy
- *Correspondence: Jacopo C. DiFrancesco
| | - Angelo Labate
- Neurophysiopathology Unit, Department of Biomedical and Dental Sciences, Morphological and Functional Images (BIOMORF), University of Messina, Messina, Italy
| | - Michele Romoli
- Section of Neurology, S. Maria della Misericordia Hospital, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Elena Chipi
- Section of Neurology, S. Maria della Misericordia Hospital, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Nicola Salvadori
- Section of Neurology, S. Maria della Misericordia Hospital, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Daniela Perani
- Nuclear Medicine Unit and Division of Neuroscience, San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Carlo Ferrarese
- Department of Neurology, ASST S. Gerardo Hospital, School of Medicine and Surgery and Milan Center for Neuroscience, University of Milano - Bicocca, Monza, Italy
| | - Cinzia Costa
- Section of Neurology, S. Maria della Misericordia Hospital, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- Cinzia Costa
| |
Collapse
|
82
|
Scherlek AA, Kozberg MG, Nicoll JAR, Perosa V, Freeze WM, van der Weerd L, Bacskai BJ, Greenberg SM, Frosch MP, Boche D, van Veluw SJ. Histopathological correlates of haemorrhagic lesions on ex vivo magnetic resonance imaging in immunized Alzheimer's disease cases. Brain Commun 2022; 4:fcac021. [PMID: 35224489 PMCID: PMC8870423 DOI: 10.1093/braincomms/fcac021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/31/2021] [Accepted: 02/01/2022] [Indexed: 12/14/2022] Open
Abstract
Haemorrhagic amyloid-related imaging abnormalities on MRI are frequently observed adverse events in the context of amyloid β immunotherapy trials in patients with Alzheimer's disease. The underlying histopathology and pathophysiological mechanisms of haemorrhagic amyloid-related imaging abnormalities remain largely unknown, although coexisting cerebral amyloid angiopathy may play a key role. Here, we used ex vivo MRI in cases that underwent amyloid β immunotherapy during life to screen for haemorrhagic lesions and assess underlying tissue and vascular alterations. We hypothesized that these lesions would be associated with severe cerebral amyloid angiopathy. Ten cases were selected from the long-term follow-up study of patients who enrolled in the first clinical trial of active amyloid β immunization with AN1792 for Alzheimer's disease. Eleven matched non-immunized Alzheimer's disease cases from an independent brain brank were used as 'controls'. Formalin-fixed occipital brain slices were imaged at 7 T MRI to screen for haemorrhagic lesions (i.e. microbleeds and cortical superficial siderosis). Samples with and without haemorrhagic lesions were cut and stained. Artificial intelligence-assisted quantification of amyloid β plaque area, cortical and leptomeningeal cerebral amyloid angiopathy area, the density of iron and calcium positive cells and reactive astrocytes and activated microglia was performed. On ex vivo MRI, cortical superficial siderosis was observed in 5/10 immunized Alzheimer's disease cases compared with 1/11 control Alzheimer's disease cases (κ = 0.5). On histopathology, these areas revealed iron and calcium positive deposits in the cortex. Within the immunized Alzheimer's disease group, areas with siderosis on MRI revealed greater leptomeningeal cerebral amyloid angiopathy and concentric splitting of the vessel walls compared with areas without siderosis. Moreover, greater density of iron-positive cells in the cortex was associated with lower amyloid β plaque area and a trend towards increased post-vaccination antibody titres. This work highlights the use of ex vivo MRI to investigate the neuropathological correlates of haemorrhagic lesions observed in the context of amyloid β immunotherapy. These findings suggest a possible role for cerebral amyloid angiopathy in the formation of haemorrhagic amyloid-related imaging abnormalities, awaiting confirmation in future studies that include brain tissue of patients who received passive immunotherapy against amyloid β with available in vivo MRI during life.
Collapse
Affiliation(s)
- Ashley A. Scherlek
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Mariel G. Kozberg
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA,J. Philip Kistler Stroke Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - James A. R. Nicoll
- Clinical Neurosciences, Clinical and Experimental Sciences School, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Valentina Perosa
- J. Philip Kistler Stroke Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Whitney M. Freeze
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Louise van der Weerd
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands,Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Brian J. Bacskai
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Steven M. Greenberg
- J. Philip Kistler Stroke Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Matthew P. Frosch
- Neuropathology Service, C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA
| | - Delphine Boche
- Clinical Neurosciences, Clinical and Experimental Sciences School, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Susanne J. van Veluw
- MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA,J. Philip Kistler Stroke Research Center, Massachusetts General Hospital/Harvard Medical School, Boston, MA, USA,Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands,Correspondence to: Susanne J. van Veluw MassGeneral Institute for Neurodegenerative Disease Massachusetts General Hospital 114 16th Street Charlestown, 02129 MA, USA E-mail:
| |
Collapse
|
83
|
Finze A, Wahl H, Janowitz D, Buerger K, Linn J, Rominger A, Stöcklein S, Bartenstein P, Wollenweber FA, Catak C, Brendel M. Regional Associations of Cortical Superficial Siderosis and β-Amyloid-Positron-Emission-Tomography Positivity in Patients With Cerebral Amyloid Angiopathy. Front Aging Neurosci 2022; 13:786143. [PMID: 35185518 PMCID: PMC8851392 DOI: 10.3389/fnagi.2021.786143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/20/2021] [Indexed: 11/20/2022] Open
Abstract
Objective This is a cross-sectional study to evaluate whether β-amyloid-(Aβ)-PET positivity and cortical superficial siderosis (cSS) in patients with cerebral amyloid angiopathy (CAA) are regionally colocalized. Methods Ten patients with probable or possible CAA (73.3 ± 10.9 years, 40% women) underwent MRI examination with a gradient-echo-T2*-weighted-imaging sequence to detect cSS and 18F-florbetaben PET examination to detect fibrillar Aβ. In all cortical regions of the Hammers Atlas, cSS positivity (MRI: ITK-SNAP segmentation) and Aβ-PET positivity (PET: ≥ mean value + 2 standard deviations of 14 healthy controls) were defined. Regional agreement of cSS- and Aβ-PET positivity was evaluated. Aβ-PET quantification was compared between cSS-positive and corresponding contralateral cSS-negative atlas regions. Furthermore, the Aβ-PET quantification of cSS-positive regions was evaluated in voxels close to cSS and in direct cSS voxels. Results cSS- and Aβ-PET positivity did not indicate similarity of their regional patterns, despite a minor association between the frequency of Aβ-positive patients and the frequency of cSS-positive patients within individual regions (rs = 0.277, p = 0.032). However, this association was driven by temporal regions lacking cSS- and Aβ-PET positivity. When analyzing all composite brain regions, Aβ-PET values in regions close to cSS were significantly higher than in regions directly affected with cSS (p < 0.0001). However, Aβ-PET values in regions close to cSS were not different when compared to corresponding contralateral cSS-negative regions (p = 0.603). Conclusion In this cross-sectional study, cSS and Aβ-PET positivity did not show regional association in patients with CAA and deserve further exploitation in longitudinal designs. In clinical routine, a specific cross-sectional evaluation of Aβ-PET in cSS-positive regions is probably not useful for visual reading of Aβ-PETs in patients with CAA.
Collapse
Affiliation(s)
- Anika Finze
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Hannes Wahl
- Department of Neuroradiology, University Hospital of Dresden, Carl Gustav Carus University Dresden, Dresden, Germany
| | - Daniel Janowitz
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Katharina Buerger
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Jennifer Linn
- Department of Neuroradiology, University Hospital of Dresden, Carl Gustav Carus University Dresden, Dresden, Germany
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sophia Stöcklein
- Department of Radiology, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Peter Bartenstein
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Frank Arne Wollenweber
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Cihan Catak
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany
| | - Matthias Brendel
- Department of Nuclear Medicine, University Hospital of Munich, LMU Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- *Correspondence: Matthias Brendel,
| |
Collapse
|
84
|
Golde TE. Disease-Modifying Therapies for Alzheimer's Disease: More Questions than Answers. Neurotherapeutics 2022; 19:209-227. [PMID: 35229269 PMCID: PMC8885119 DOI: 10.1007/s13311-022-01201-2] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/09/2022] [Indexed: 12/17/2022] Open
Abstract
Scientific advances over the last four decades have steadily infused the Alzheimer's disease (AD) field with great optimism that therapies targeting Aβ, amyloid, tau, and innate immune activation states in the brain would provide disease modification. Unfortunately, this optimistic scenario has not yet played out. Though a recent approval of the anti-Aβ aggregate binding antibody, Aduhelm (aducanumab), as a "disease-modifying therapy for AD" is viewed by some as a breakthrough, many remain unconvinced by the data underlying this approval. Collectively, we have not succeeded in changing AD from a largely untreatable, inevitable, and incurable disease to a treatable, preventable, and curable one. Here, I will review the major foci of the AD "disease-modifying" therapeutic pipeline and some of the "open questions" that remain in terms of these therapeutic approaches. I will conclude the review by discussing how we, as a field, might adjust our approach, learning from our past failures to ensure future success.
Collapse
Affiliation(s)
- Todd E Golde
- Departments of Neuroscience and Neurology, Center for Translational Research in Neurodegenerative Disease, Evelyn F. and William L. McKnight Brain Institute, Norman Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
85
|
A case of cerebral amyloid angiopathy related inflammation after vaccination against SARS-CoV-2. NEUROIMMUNOLOGY REPORTS 2022. [PMCID: PMC9308493 DOI: 10.1016/j.nerep.2022.100120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Background Cerebral amyloid angiopathy related inflammation (CAA-RI) is a neuroinflammatory disease that is associated with perivascular amyloid- deposition. Case presentation A middle-aged woman with a remote history of autoimmune disorders presented with unilateral migraine headaches, dizziness, unsteadiness, and fogginess 36 hours after administration of mRNA vaccine against SARS-CoV-2. Initially, unilateral leptomeningeal enhancement on MRI on the same side of headaches raised suspicion for leptomeningeal involvement of her known cutaneous T-cell lymphoma in remission. After two relatively unremarkable CSF analyses, she underwent a brain biopsy which showed amyloid deposits in vessels instead of lymphomatous infiltration. She was diagnosed with CAA-RI, and the headache and cognitive symptoms responded well to high-dose corticosteroids with a slow taper. Discussion/conclusion We review the clinical literature of CAA-RI and its potential association with amyloid-related imaging abnormalities (ARIA) after administration of immunotherapy against amyloid.
Collapse
|
86
|
Aldea R, Grimm HP, Gieschke R, Hofmann C, Lott D, Bullain S, Delmar P, Klein G, Lyons M, Piazza F, Carare RO, Mazer NA. In silico exploration of amyloid‐related imaging abnormalities in the gantenerumab open‐label extension trials using a semi‐mechanistic model. ALZHEIMER'S & DEMENTIA: TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2022; 8:e12306. [PMID: 35676943 PMCID: PMC9169977 DOI: 10.1002/trc2.12306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/12/2022] [Accepted: 04/25/2022] [Indexed: 11/28/2022]
Abstract
Introduction Amyloid‐related imaging abnormalities with edema/effusion (ARIA‐E) are commonly observed with anti‐amyloid therapies in Alzheimer's disease. We developed a semi‐mechanistic, in silico model to understand the time course of ARIA‐E and its dose dependency. Methods Dynamic and statistical analyses of data from 112 individuals that experienced ARIA‐E in the open‐label extension of SCarlet RoAD (a study of gantenerumab in participants with prodromal Alzheimer's disease) and Marguerite RoAD (as study of Gantenerumab in participants with mild Alzheimer's disease) studies were used for model building. Gantenerumab pharmacokinetics, local amyloid removal, disturbance and repair of the vascular wall, and ARIA‐E magnitude were represented in the novel vascular wall disturbance (VWD) model of ARIA‐E. Results The modeled individual‐level profiles provided a good representation of the observed pharmacokinetics and time course of ARIA‐E magnitude. ARIA‐E dynamics were shown to depend on the interplay between drug‐mediated amyloid removal and intrinsic vascular repair processes. Discussion Upon further refinement and validation, the VWD model could inform strategies for dosing and ARIA monitoring in individuals with an ARIA‐E history.
Collapse
Affiliation(s)
- Roxana Aldea
- Roche Pharma Research and Early Development Roche Innovation Center Basel Switzerland
| | - Hans Peter Grimm
- Roche Pharma Research and Early Development Roche Innovation Center Basel Switzerland
| | - Ronald Gieschke
- Roche Pharma Research and Early Development Roche Innovation Center Basel Switzerland
| | - Carsten Hofmann
- Roche Pharma Research and Early Development Roche Innovation Center Basel Switzerland
| | - Dominik Lott
- Roche Pharma Research and Early Development Roche Innovation Center Basel Switzerland
| | - Szofia Bullain
- Roche Product Development Neuroscience Basel Switzerland
| | - Paul Delmar
- Roche Product Development Neuroscience Basel Switzerland
| | - Gregory Klein
- Roche Pharma Research and Early Development Roche Innovation Center Basel Switzerland
| | | | - Fabrizio Piazza
- School of Medicine Laboratory of CAA and AD Translational Research and Biomarkers University of Milano‐Bicocca Monza Italy
| | - Roxana O. Carare
- Faculty of Medicine Interdisciplinary Dementia and Aging Centre University of Southampton Southampton UK
| | - Norman A. Mazer
- Roche Pharma Research and Early Development Roche Innovation Center Basel Switzerland
| |
Collapse
|