251
|
Chitnis T, Tenembaum S, Banwell B, Krupp L, Pohl D, Rostasy K, Yeh EA, Bykova O, Wassmer E, Tardieu M, Kornberg A, Ghezzi A. Consensus statement: evaluation of new and existing therapeutics for pediatric multiple sclerosis. Mult Scler 2011; 18:116-27. [DOI: 10.1177/1352458511430704] [Citation(s) in RCA: 143] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
New therapies are being evaluated by clinical trials and, if efficacious, introduced for the treatment of adult MS. The role of these new and existing agents in the management of pediatric MS has yet to be defined. Pediatric investigation plans are now required by the Food and Drug Administration and European Medicines Agency for approval of new biological agents, providing an important opportunity to gather much-needed data for clinicians caring for children and adolescents with MS. However, challenges include the small number of patients, and the need for efficient yet comprehensive study designs incorporating factors necessary to inform the clinical care of children with MS. The elected Steering committee of the International Pediatric MS Study Group (IPMSSG) conducted a structured review of existing data on the disease-modifying therapies in pediatric MS and developed a consensus statement, which was further modified by the IPMSSG general membership, using an online survey tool. Fifty-one IPMSSG members from 21 countries responded to the survey, and 50 approved the final statement. Consensus recommendations regarding use of existing first- and second-line therapies, as well as a proposed definition for inadequate treatment response, are presented. Recommendations for the use and evaluation of emerging therapies (currently in phase III clinical trials or recently approved for adult MS) are discussed. The IPMSSG endorses the inclusion of pediatric MS patients in trials evaluating appropriate new and emerging therapies. Mechanisms for conducting high-impact, multicenter studies, including long-term follow-up in pediatric MS, are required to ensure that all MS patients, irrespective of age, benefit from advances in MS therapeutics.
Collapse
Affiliation(s)
- T Chitnis
- Partners Pediatric Multiple Sclerosis Center, Massachusetts General Hospital, Boston, MA, USA
| | - S Tenembaum
- National Pediatric Hospital, Dr J P Garrahan, Buenos Aires, Argentina
| | - B Banwell
- The Hospital for Sick Children, University of Toronto, Canada
| | - L Krupp
- Stony Brook University Medical Center, Stony Brook, NY, USA
| | - D Pohl
- Children’s Hospital of Eastern Ontario, University of Ottawa, Canada
| | - K Rostasy
- Department of Pediatrics IV, Division of Pediatric Neurology and Inborn Errors of Metabolism, Innsbruck Medical University, Innsbruck, Austria
| | - E A Yeh
- Department of Neurology, SUNY Buffalo, Buffalo, NY, USA
| | - O Bykova
- Moscow Pediatric Psychoneurological Hospital, Moscow, Russia
| | - E Wassmer
- Birmingham Children’s Hospital, Birmingham, UK
| | - M Tardieu
- Assistance Publique-Hôpitaux de Paris, Hôpital Bicêtre and Université Paris-Sud, Paris, France
| | - A Kornberg
- Royal Children’s Hospital, Melbourne, Australia
| | - A Ghezzi
- Multiple Sclerosis Study Center, Hospital of Gallarate, Gallarate, Italy
| | | |
Collapse
|
252
|
Sahraian MA, Radue EW, Eshaghi A, Besliu S, Minagar A. Progressive multifocal leukoencephalopathy: a review of the neuroimaging features and differential diagnosis. Eur J Neurol 2011; 19:1060-9. [PMID: 22136455 DOI: 10.1111/j.1468-1331.2011.03597.x] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Progressive multifocal leukoencephalopathy (PML) is an uncommon and often fatal demyelinating disease of human central nervous system, which is caused by reactivation of the polyomavirus JC (JCV). PML generally occurs in patients with profound immunosuppression such as AIDS patients. Recently, a number of PML cases have been associated with administration of natalizumab for treatment of multiple sclerosis (MS) patients. Diagnosis and management of PML became a major concern after its occurrence in multiple sclerosis patients treated with natalizumab. Diagnosis of PML usually rests on neuroimaging in the appropriate clinical context and is further confirmed by cerebrospinal fluid polymerase chain reaction (PCR) for JCV DNA. Treatment with antiretroviral therapies in HIV-seropositive patients or discontinuing natalizumab in MS patients with PML may lead to the development of immune reconstitution inflammatory syndrome (IRIS) which presents with deterioration of the previous symptoms and may lead to death. In patients under treatment with monoclonal antibodies in routine practice, or new ones in ongoing clinical trials, differentiating PML from new MS lesions on brain MRI is critical for both the neurologists and neuroradiologists. In this review, we discuss the clinical features, neuroimaging manifestations of PML, IRIS and neuroimaging clues to differentiate new MS lesions from PML. In addition, various neuroimaging features of PML on the non-conventional MR techniques such as diffusion-weighted imaging (DWI), diffusion tensor imaging (DTI), and MR spectroscopy (MRS) are discussed.
Collapse
Affiliation(s)
- M A Sahraian
- Sina MS Research Center, Brain and Spinal Injury Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | | | | | | | | |
Collapse
|
253
|
Aringer M, Burkhardt H, Burmester GR, Fischer-Betz R, Fleck M, Graninger W, Hiepe F, Jacobi AM, Kötter I, Lakomek HJ, Lorenz HM, Manger B, Schett G, Schmidt RE, Schneider M, Schulze-Koops H, Smolen JS, Specker C, Stoll T, Strangfeld A, Tony HP, Villiger PM, Voll R, Witte T, Dörner T. Current state of evidence on 'off-label' therapeutic options for systemic lupus erythematosus, including biological immunosuppressive agents, in Germany, Austria and Switzerland--a consensus report. Lupus 2011; 21:386-401. [PMID: 22072024 DOI: 10.1177/0961203311426569] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Systemic lupus erythematosus (SLE) can be a severe and potentially life-threatening disease that often represents a therapeutic challenge because of its heterogeneous organ manifestations. Only glucocorticoids, chloroquine and hydroxychloroquine, azathioprine, cyclophosphamide and very recently belimumab have been approved for SLE therapy in Germany, Austria and Switzerland. Dependence on glucocorticoids and resistance to the approved therapeutic agents, as well as substantial toxicity, are frequent. Therefore, treatment considerations will include 'off-label' use of medication approved for other indications. In this consensus approach, an effort has been undertaken to delineate the limits of the current evidence on therapeutic options for SLE organ disease, and to agree on common practice. This has been based on the best available evidence obtained by a rigorous literature review and the authors' own experience with available drugs derived under very similar health care conditions. Preparation of this consensus document included an initial meeting to agree upon the core agenda, a systematic literature review with subsequent formulation of a consensus and determination of the evidence level followed by collecting the level of agreement from the panel members. In addition to overarching principles, the panel have focused on the treatment of major SLE organ manifestations (lupus nephritis, arthritis, lung disease, neuropsychiatric and haematological manifestations, antiphospholipid syndrome and serositis). This consensus report is intended to support clinicians involved in the care of patients with difficult courses of SLE not responding to standard therapies by providing up-to-date information on the best available evidence.
Collapse
Affiliation(s)
- M Aringer
- Rheumatology, Medicine III, University Medical Center TU Dresden, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
254
|
Kato H, Yamamoto K, Taji H, Oki Y, Chihara D, Seto M, Kagami Y, Morishima Y. Interstitial pneumonia after autologous hematopoietic stem cell transplantation in B-cell non-hodgkin lymphoma. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2011; 11:483-9. [PMID: 21978956 DOI: 10.1016/j.clml.2011.06.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2011] [Revised: 06/01/2011] [Accepted: 06/15/2011] [Indexed: 11/25/2022]
Abstract
INTRODUCTION During the past decade, interstitial pneumonia (IP) is one of the newly recognized adverse events regarding rituximab therapy. However, disease characteristics of IP after autologous hematopoietic stem cell transplantation (ASCT) have not been well-described since the introduction of rituximab. PATIENTS AND METHODS We retrospectively analyzed 103 patients with B-cell non-Hodgkin lymphoma undergoing ASCT. A propensity scoring system was applied in our analysis to eliminate potential confounding factors of covariates. RESULTS The total number of patients who developed IP was nine. Five patients developed IP among 57 patients previously treated with rituximab, and four patients developed IP among 46 who were rituximab-naïve. Cumulative incidence of IP was 7.8% at 1 year. Among the patients using rituximab, one patient had IP during the peri-engraftment period (cytomegalovirus infection), three patients had IP between 3 and 12 months (Pneumocystis pneumonia [PCP, n = 1] and unknown cause [n = 2]), and the other one patient had IP 3.3 years after ASCT (unknown cause). Four patients in the rituximab-naïve group developed IP between 3 and 12 months (PCP [n = 1] and unknown cause [n = 3]). All nine patients had symptomatic episodes before IP, three of which died of IP or secondary infections. Patients receiving a total body irradiation conditioning regimen had a higher risk of IP (odds ratio = 3.6, P < .001), whereas the incidence was not affected by rituximab usage (P = .85, Log-rank test). CONCLUSION This study shows that the rituximab usage was not identified as a risk factor of IP and that total body irradiation was the only independent risk factor for IP. Close monitoring is encouraged when symptomatic unexplained episodes are identified during follow-up examinations after ASCT.
Collapse
Affiliation(s)
- Harumi Kato
- Department of Hematology and Cell Therapy, Aichi Cancer Center Hospital, Chikusa-ku, Nagoya, Japan
| | | | | | | | | | | | | | | |
Collapse
|
255
|
LFA-1 blockade induces effector and regulatory T-cell enrichment in lymph nodes and synergizes with CTLA-4Ig to inhibit effector function. Blood 2011; 118:5851-61. [PMID: 21972294 DOI: 10.1182/blood-2011-04-347252] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Despite encouraging results using lymphocyte function antigen-1 (LFA-1) blockade to inhibit BM and solid organ transplantation rejection in nonhuman primates and humans, the precise mechanisms underlying its therapeutic potential are still poorly understood. Using a fully allogeneic murine transplantation model, we assessed the relative distribution of total lymphocyte subsets in untreated versus anti-LFA-1-treated animals. Our results demonstrated a striking loss of naive T cells from peripheral lymph nodes, a concomitant gain in blood after LFA-1 blockade, and a shift in phenotype of the cells remaining in the node to a CD62LloCD44hi profile. We determined that this change was due to a specific enrichment of activated, graft-specific effectors in the peripheral lymph nodes of anti-LFA-1-treated mice compared with untreated controls, and not to a direct effect of anti-LFA-1 on CD62L expression. LFA-1 blockade also resulted in a dramatic increase in the frequency of CD4+ FoxP3+ regulatory T cells in graft-draining nodes. Our results suggest that the differential impact of LFA-1 blockade on the distribution of naive versus effector and regulatory T cells may underlie its ability to inhibit alloreactive T-cell responses after transplantation.
Collapse
|
256
|
Pitini V, Arrigo C, Sauta MG, Altavilla G. Risk of second malignant neoplasm among patients with lymphoma. J Clin Oncol 2011; 29:3834; author reply 3834-5. [PMID: 21876085 DOI: 10.1200/jco.2011.37.2144] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
257
|
Kappos L, Bates D, Edan G, Eraksoy M, Garcia-Merino A, Grigoriadis N, Hartung HP, Havrdová E, Hillert J, Hohlfeld R, Kremenchutzky M, Lyon-Caen O, Miller A, Pozzilli C, Ravnborg M, Saida T, Sindic C, Vass K, Clifford DB, Hauser S, Major EO, O'Connor PW, Weiner HL, Clanet M, Gold R, Hirsch HH, Radü EW, Sørensen PS, King J. Natalizumab treatment for multiple sclerosis: updated recommendations for patient selection and monitoring. Lancet Neurol 2011; 10:745-58. [PMID: 21777829 DOI: 10.1016/s1474-4422(11)70149-1] [Citation(s) in RCA: 195] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Natalizumab, a highly specific α4-integrin antagonist, is approved for treatment of patients with active relapsing-remitting multiple sclerosis (RRMS). It is generally recommended for individuals who have not responded to a currently available first-line disease-modifying therapy or who have very active disease. The expected benefits of natalizumab treatment have to be weighed against risks, especially the rare but serious adverse event of progressive multifocal leukoencephalopathy. In this Review, we revisit and update previous recommendations on natalizumab for treatment of patients with RRMS, based on additional long-term follow-up of clinical studies and post-marketing observations, including appropriate patient selection and management recommendations.
Collapse
|
258
|
Beyond the joints: neurological involvement in rheumatoid arthritis. Clin Rheumatol 2011; 31:1-12. [PMID: 21932019 DOI: 10.1007/s10067-011-1841-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 08/28/2011] [Indexed: 10/17/2022]
Abstract
Although arthritis is the most notable component, rheumatoid arthritis (RA) is a systemic inflammatory disorder where extra-articular manifestations are common; among them, central and peripheral nervous system involvement is frequent and associated with significant morbidity and, in some cases, reduced life span. It may produce a myriad of symptoms and signs ranging from subtle numbness in a hand, to quadriparesis and sudden death. Central and peripheral neurologic manifestations may arise from structural damage produced by RA in diarthroidal joints, by the systemic inflammatory process of the disease itself or by the drugs used to treat it. Neurologic syndromes may appear suddenly or developed slowly through months, and emerge early or after years of having RA. Neurologic manifestations may be easily overlooked or incorrectly assigned to peripheral arthritis unless the attending physician is aware of these complications. In this article, we review neurologic involvement in RA patients with emphasis on clinical approach for early detection.
Collapse
|
259
|
|
260
|
Uettwiller F, Rigal E, Hoarau C. Infections associated with monoclonal antibody and fusion protein therapy in humans. MAbs 2011; 3:461-6. [PMID: 21822056 DOI: 10.4161/mabs.3.5.16553] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Monoclonal antibodies (mAbs), especially those that interact with immune or hematologic leukocyte membrane targets, have changed the outcome of numerous diseases. However, mAbs can block or reduce immune cells and cytokines, and can lead to increased risk of infection. Some of these risks are predictable and can be explained by their mechanisms of action. Others have been observed only after the mAbs were licensed and used extensively in patients. In this review, we focus on infectious complications that occur upon treatment with mAbs or Fc-containing fusion proteins targeting leukocyte membrane proteins, including CD52, CD20, tumor necrosis factor, VLA4, CD11a and CTLA4. We report their known infectious risks and the recommendations for their use. Although most of these drugs are clinically safe when the indications are respected, we emphasize the need for regular updating of pharmacovigilance data.
Collapse
Affiliation(s)
- Florence Uettwiller
- Allergology and Clinical Immunology Unit, Pediatric Unit, Clocheville Hospital, "CDIG" François Rabelais University, Tours, France
| | | | | |
Collapse
|
261
|
The London position statement of the World Congress of Gastroenterology on Biological Therapy for IBD with the European Crohn's and Colitis Organisation: safety. Am J Gastroenterol 2011; 106:1594-602; quiz 1593, 1603. [PMID: 21844919 DOI: 10.1038/ajg.2011.211] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This paper in the series from the World Congress of Gastroenterology addresses the safety and immunogenicity of biological therapy. The safety profile in randomized controlled studies of all biological agents in Crohn's disease (CD) and ulcerative colitis has been generally favorable, but a small percentage of patients experience severe side effects on biological therapy, including pneumonia, tuberculosis, lymphoma, demyelination, drug-induced lupus, or hepatotoxicity. Although there is unequivocal evidence of an increased risk of serious infection among patients with rheumatoid arthritis treated with anti-tumor necrosis factor therapy, the evidence is less clear in CD. The risk of infection may be increased by combination therapy with steroids and/or immunomodulators. There is a specific risk of the rare γ δ hepatosplenic lymphoma that appears to have a predeliction for young males on combination therapy. The α4 integrin antagonist natalizumab also carries a specific risk of progressive multifocal leucoencephalopathy and reactivation of JC virus infection. The immunogenicity of biological therapy is complex: all agents are potentially immunogenic and this can be reduced by combination with immunomodulators. This may enhance both therapeutic efficacy and the risk of infection or malignancy, so the balance of risk and benefit must be judged for individual patients.
Collapse
|
262
|
Viscidi RP, Khanna N, Tan CS, Li X, Jacobson L, Clifford DB, Nath A, Margolick JB, Shah KV, Hirsch HH, Koralnik IJ. JC virus antibody and viremia as predictors of progressive multifocal leukoencephalopathy in human immunodeficiency virus-1-infected individuals. Clin Infect Dis 2011; 53:711-5. [PMID: 21852452 DOI: 10.1093/cid/cir507] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
We examined whether prediagnostic John Cunningham virus (JCV) antibodies and viremia are predictors of progressive multifocal leukoencephalopathy (PML) in 83 PML cases and 240 human immunodeficiency virus (HIV) disease-matched controls. JCV viremia was not predictive of PML, but some patients showed higher anti-JCV immunoglobulin G (IgG) responses 6 months prior to diagnosis.
Collapse
Affiliation(s)
- Raphael P Viscidi
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
263
|
Tan IL, McArthur JC, Clifford DB, Major EO, Nath A. Immune reconstitution inflammatory syndrome in natalizumab-associated PML. Neurology 2011; 77:1061-7. [PMID: 21832229 DOI: 10.1212/wnl.0b013e31822e55e7] [Citation(s) in RCA: 160] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To study the outcome of patients with multiple sclerosis (MS) and with natalizumab-associated progressive multifocal leukoencephalopathy (PML) and immune reconstitution inflammatory syndrome (IRIS). METHODS MedWatch reports from Biogen-Idec (manufacturer of natalizumab, Tysabri(®)) were reviewed which comprised all 42 cases of natalizumab-related PML cases since its reintroduction until March 2010. RESULTS All except 2 patients with natalizumab-related PML were managed by discontinuation of natalizumab and plasmapheresis/immunoadsorption (PLEX/IA). Seventeen patients had contrast enhancement of PML lesions on neuroimaging at the time of diagnosis before withdrawal/removal of natalizumab (early-PML-IRIS) and 23 patients developed contrast enhancement only after withdrawal/removal of natalizumab (late-PML-IRIS). All patients developed IRIS. IRIS was defined as worsening of neurologic deficits during the immune reconstitution following discontinuation of natalizumab, corroborated by inflammatory changes on neuroimaging. Following PLEX/IA, JC viral load in CSF increased by >10 fold in those with early-PML-IRIS but <2 fold in late-PML-IRIS. IRIS developed earlier and was more severe in early-PML-IRIS (p < 0.05). At the last follow-up, all patients had worse EDSS scores but this was higher in patients with early-PML-IRIS compared to those with late-PML-IRIS (p > 0.05). Mortality was comparable between the 2 groups, 29.4 ± 11% vs 21.7 ± 8.8%. Corticosteroid therapy during IRIS was associated with better Expanded Disability Status Scale outcome, p < 0.05. CONCLUSION Early immunologic rebound in natalizumab-associated PML has worse survival and neurologic outcome. PLEX/IA may accelerate IRIS and its impact on the final outcome is unclear. Corticosteroid therapy provides a modest benefit and needs to be systemically studied in a controlled manner in the management of natalizumab-associated PML-IRIS.
Collapse
Affiliation(s)
- I L Tan
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | | | | | | | | |
Collapse
|
264
|
Khan A, Scott D. Rituximab after methotrexate failure in rheumatoid arthritis: evaluation of the SERENE trial. Expert Opin Biol Ther 2011; 11:1515-8. [DOI: 10.1517/14712598.2011.608061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
265
|
Sellier-Leclerc AL, Baudouin V, Kwon T, Macher MA, Guérin V, Lapillonne H, Deschênes G, Ulinski T. Rituximab in steroid-dependent idiopathic nephrotic syndrome in childhood--follow-up after CD19 recovery. Nephrol Dial Transplant 2011; 27:1083-9. [PMID: 21810762 DOI: 10.1093/ndt/gfr405] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Anne-Laure Sellier-Leclerc
- Department of Pediatric Nephrology, Robert Debré Hospital, Assistance Publique Hopitaux de Paris, Paris, France.
| | | | | | | | | | | | | | | |
Collapse
|
266
|
Plasma HHV8 DNA predicts relapse in individuals with HIV-associated multicentric Castleman disease. Blood 2011; 118:271-5. [DOI: 10.1182/blood-2011-02-335620] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Abstract
HIV-associated multicentric Castleman disease (HIV-MCD) is a rare lymphoproliferative disorder caused by infection with human herpesvirus-8. The disease follows a relapsing and remitting clinical course, with marked systemic symptoms during an active attack, which can prove fatal. Its incidence is rising, and new data indicate the utility of the anti-CD20 monoclonal antibody rituximab at inducing remissions in both first- and second-line settings, although biomarkers associated with relapse have not been previously identified. In 52 individuals with a histologic diagnosis of HIV-MCD, we performed univariate and multivariate analyses to predict factors associated with an HIV-MCD attack. Although a younger age (< 50 years) was associated with an attack, the strongest association was observed with plasma levels of human herpesvirus-8 DNA. Rising levels predicted an attack (hazard ratio = 2.9; 95% confidence interval, 1.3-6.7), and maintenance therapy with rituximab should be considered in these individuals.
Collapse
|
267
|
Wu J, Langford LA, Schellingerhout D, Guha-Thakurta N, Tummala S, Weinberg JS, Puduvalli VK. Progressive multifocal leukoencephalopathy in a patient with glioblastoma. J Neurooncol 2011; 103:791-6. [PMID: 21120583 PMCID: PMC5546753 DOI: 10.1007/s11060-010-0453-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Accepted: 10/25/2010] [Indexed: 02/03/2023]
Abstract
Malignant gliomas are aggressive malignancies which inevitably recur despite multimodality treatment. In a subset of patients who are longer term survivors of this disease, progressive radiologic worsening can also occur from late effects of radiation rather than recurrent tumor, a differential diagnosis that is commonly considered in this setting. However, other causes for radiologic progression are not as well recognized and could potentially confound management leading to incorrect treatment decisions. Progressive multifocal leukoencephalopathy (PML) is a rare infectious demyelinating disease of the central nervous system seen primarily in immunocompromised patients, the early diagnosis and treatment of which remains a challenge. Here, we report a case of a long term survivor with glioblastoma whose diagnostic and therapeutic management was confounded by the development of PML. We review the radiological features and clinical course of this patient to highlight the dramatic neurological course in the setting of a highly malignant tumor, and emphasize the unusual changes in diffusion weighted images, and the need for clinical suspicion for early diagnosis of PML.
Collapse
Affiliation(s)
- Jing Wu
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lauren A. Langford
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Dawid Schellingerhout
- Department of Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Nandita Guha-Thakurta
- Department of Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sudhakar Tummala
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jeffrey S. Weinberg
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Vinay K. Puduvalli
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
268
|
Natalizumab para la esclerosis múltiple remitente-recurrente. Neurologia 2011; 26:357-68. [DOI: 10.1016/j.nrl.2010.10.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2010] [Accepted: 06/16/2010] [Indexed: 11/20/2022] Open
|
269
|
Progressive multifocal leukoencephalopathy and natalizumab. J Neurol 2011; 258:1920-8. [PMID: 21647730 DOI: 10.1007/s00415-011-6116-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 05/18/2011] [Accepted: 05/20/2011] [Indexed: 01/09/2023]
Abstract
Natalizumab (TYSABRI(®)), a specific α4-integrin antagonist, is approved as a second-line treatment of relapsing-remitting MS (RRMS) patients who fail therapy with interferons or as first-line treatment of patients with highly active relapsing-remitting disease. Since the market introduction of natalizumab as a monotherapy in July of 2006, 111 cases of PML have been reported in natalizumab-treated MS patients as of April 2011. This review focuses on the available data regarding risk stratification for PML under long-term natalizumab therapy, and summarizes the current approach for PML management, as a natalizumab treatment complication is not necessarily associated with a fatal outcome. There is a need for development of surrogate markers that would help to better define the risk of PML in individual patients.
Collapse
|
270
|
|
271
|
Marie I, Guegan-Massardier E, Levesque H. Progressive multifocal leukoencephalopathy in refractory polymyositis treated with rituximab. Eur J Intern Med 2011; 22:e13-4. [PMID: 21570627 DOI: 10.1016/j.ejim.2011.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2010] [Revised: 12/23/2010] [Accepted: 01/07/2011] [Indexed: 10/18/2022]
|
272
|
Buch MH, Smolen JS, Betteridge N, Breedveld FC, Burmester G, Dörner T, Ferraccioli G, Gottenberg JE, Isaacs J, Kvien TK, Mariette X, Martin-Mola E, Pavelka K, Tak PP, van der Heijde D, van Vollenhoven RF, Emery P, for the Rituximab Consensus Expert Committee. Updated consensus statement on the use of rituximab in patients with rheumatoid arthritis. Ann Rheum Dis 2011; 70:909-20. [PMID: 21378402 PMCID: PMC3086093 DOI: 10.1136/ard.2010.144998] [Citation(s) in RCA: 325] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Collaborators] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2011] [Indexed: 01/19/2023]
Abstract
BACKGROUND Since initial approval for the treatment of rheumatoid arthritis (RA), rituximab has been evaluated in clinical trials involving various populations with RA. Information has also been gathered from registries. This report therefore updates the 2007 consensus document on the use of rituximab in the treatment of RA. METHODS Preparation of this new document involved many international experts experienced in the treatment of RA. Following a meeting to agree upon the core agenda, a systematic literature review was undertaken to identify all relevant data. Data were then interrogated by a drafting committee, with subsequent review and discussion by a wider expert committee leading to the formulation of an updated consensus statement. These committees also included patients with RA. RESULTS The new statement covers wide-ranging issues including the use of rituximab in earlier RA and impact on structural progression, and aspects particularly pertinent to rituximab such as co-medication, optimal dosage regimens, repeat treatment cycles and how to manage non-response. Biological therapy following rituximab usage is also addressed, and safety concerns including appropriate screening for hepatitis, immunoglobulin levels and infection risk. This consensus statement will support clinicians and inform patients when using B-cell depletion in the management of RA, providing up-to-date information and highlighting areas for further research. CONCLUSION New therapeutic strategies and treatment options for RA, a chronic destructive and disabling disease, have expanded over recent years. These have been summarised in general strategic suggestions and specific management recommendations, emphasising the importance of expedient disease-modifying antirheumatic drug implementation and tight disease control. This consensus statement is in line with these fundamental principles of management.
Collapse
Affiliation(s)
- Maya H Buch
- Section of Musculoskeletal Diseases, Leeds Institute of Molecular Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Josef S Smolen
- Department of Internal Medicine 3, Division of Rheumatology, Medical University of Vienna, Vienna, Austria
- Center for Rheumatic Diseases, Hietzing Hospital, Vienna, Austria
| | | | - Ferdinand C Breedveld
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gerd Burmester
- Department of Rheumatology and Clinical Immunology, Humboldt University, Charite Hospital, Berlin, Germany
| | - Thomas Dörner
- Department of Rheumatology and Clinical Immunology, Humboldt University, Charite Hospital, Berlin, Germany
| | | | - Jacques-Eric Gottenberg
- Rheumatology Department, Hôpitaux Universitaires de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - John Isaacs
- Institute of Cellular Medicine, Newcastle University Medical School, Newcastle upon Tyne, UK
| | - Tore K Kvien
- Department of Rheumatology, Diakonhjemmet Hospital, Oslo, Norway
| | - Xavier Mariette
- Institut Pour la Santé et la Recherche Médicale (INSERM) U 1012, Hôpital Bicêtre, Université Paris-Sud 11, Paris, France
| | | | - Karel Pavelka
- Institute of Rheumatology and Clinic of Rheumatology Charles University, Prague, Czech Republic
| | - Paul P Tak
- Division of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, Amsterdam, The Netherlands
| | | | - Ronald F van Vollenhoven
- Rheumatology Unit, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Solna, Sweden
| | - Paul Emery
- Section of Musculoskeletal Diseases, Leeds Institute of Molecular Medicine, University of Leeds, Leeds, UK
- NIHR Leeds Musculoskeletal Biomedical Research Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | | |
Collapse
Collaborators
Jordi Carbonell Abello, Marwan Bukhari, Harald Burkhardt, Bernard Combe, Juan-Jesus Gomez-Reino Carnota, Leonor Barile Fabris, Lars Klareskog, Jose Luis Marenco de la Fuente, Carlo-Maurizio Montecucco, Mikkel Ostergaard, Eliseo Pascual Gomez, Raimon Sanmarti Sala, Hans-Peter Tony, Guido Valesini, Jaap van Laar, Piet van Riel,
Collapse
|
273
|
Abstract
Progressive multifocal leukoencephalopathy (PML) is a disease of the central nervous system (CNS) with destructive infection of oligodendrocytes by JC virus. PML belongs to the opportunistic infections. It is observed in patients with HIV infection, lymphoid malignancies, after organ- and stem cell transplantations and more recently in the context of modern immune-therapies with monoclonal antibodies (mAb) like natalizumab, rituximab, infliximab and efalizumab. The natural course of PML is fatal within months. More recently, the Immune Reconstitution Inflammatory Syndrome (IRIS) has been observed in patients with HIV infection treated with combination antiretroviral therapy (cART) as well as patients in whom the PML-inducing immune therapy has been terminated. In PML-IRIS the immune system contributes to the elimination of JC virus from the CNS and if PML-IRIS emerges, PML can be survived but can lead as well to catastrophic outcomes with brain herniation and death. Therefore the management of IRIS requires special knowledge in JC virus biology and patient care. JC virus infection is possibly involved in a variety of additional neurological conditions and cancer. Much will be learned within the next years that could change our view on the understanding of JC virus and human disease.
Collapse
Affiliation(s)
- Robert Weissert
- Department of Neurology, Geneva University Hospital, Micheli-du-Crest 24, 1211 Geneva 14, Switzerland.
| |
Collapse
|
274
|
Clifford DB, Ances B, Costello C, Rosen-Schmidt S, Andersson M, Parks D, Perry A, Yerra R, Schmidt R, Alvarez E, Tyler KL. Rituximab-associated progressive multifocal leukoencephalopathy in rheumatoid arthritis. ACTA ACUST UNITED AC 2011; 68:1156-64. [PMID: 21555606 DOI: 10.1001/archneurol.2011.103] [Citation(s) in RCA: 218] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To describe the development of progressive multifocal leukoencephalopathy (PML) in patients with rheumatoid arthritis (RA) treated with rituximab. DESIGN Case study. SETTING Clinical care for patients with rheumatologic diseases. Most were referred to academic centers for care after diagnosis (Washington University, St Louis, Missouri; Karolinska Insitute, Stockholm, Sweden; and Royal Melbourne Hospital, Melbourne, Australia) while one was cared for in a neurology practice in Dallas, Texas, with consultation by an academic neurovirologist from the University of Colorado in Denver. PATIENTS Four patients developing PML in the setting of rituximab therapy for RA. INTERVENTION Rituximab therapy. MAIN OUTCOME MEASURES Clinical and pathological observations. RESULTS Four patients from an estimated population of 129 000 exposed to rituximab therapy for RA are reported in whom PML developed after administration of this drug. All were women older than 50 years, commonly with Sjögren syndrome and a history of treatment for joint disease ranging from 3 to 14 years. One case had no prior biologic and minimal immunosuppressive therapy. Progressive multifocal leukoencephalopathy presented as a progressive neurological disorder, with diagnosis confirmed by detection of JC virus DNA in the cerebrospinal fluid or brain biopsy specimen. Two patients died in less than 1 year from PML diagnosis, while 2 remain alive after treatment withdrawal. Magnetic resonance scans and tissue evaluation confirmed the frequent development of inflammatory PML during the course of the disease. CONCLUSION These cases suggest an increased risk, about 1 case per 25 000 individuals, of PML in patients with RA being treated with rituximab. Inflammatory PML may occur in this setting even while CD20 counts remain low.
Collapse
Affiliation(s)
- David B Clifford
- Department of Neurology, Washington University, St Louis, MO 63110, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
275
|
van Oers MHJ, Kersten MJ. Treatment strategies in advanced stage follicular lymphoma. Best Pract Res Clin Haematol 2011; 24:187-201. [PMID: 21658618 DOI: 10.1016/j.beha.2011.03.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Although the introduction of anti-CD20 monoclonal antibodies has improved the outcome of patients with follicular lymphoma, a curative treatment is still not available. Many questions still remain to be answered: when should treatment be initiated? Is there an optimal first line treatment and can this treatment be individualized on the basis of prognostic markers? What is the best treatment strategy for relapsed follicular lymphoma and what is the place of the many novel agents? Should maintenance treatment be given to all patients and how? In the present review we will address these questions.
Collapse
Affiliation(s)
- Marinus H J van Oers
- Department of Haematology, F4-224, Academic Medical Centre, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.
| | | |
Collapse
|
276
|
Plevy SE, Targan SR. Future therapeutic approaches for inflammatory bowel diseases. Gastroenterology 2011; 140:1838-46. [PMID: 21530750 PMCID: PMC5076881 DOI: 10.1053/j.gastro.2011.02.014] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2010] [Revised: 02/04/2011] [Accepted: 02/11/2011] [Indexed: 12/13/2022]
Abstract
In this review, we speculate about future therapeutic approaches for inflammatory bowel diseases (IBDs), focusing on the need for better preclinical and clinical models and approaches beyond small molecules and systemically administered biologics. We offer ideas to change clinical trial programs and to use immunologic and genetic biomarkers to personalize medicine. We attempt to reconcile past therapeutic successes and failures to improve future approaches. Some of our ideas might be provocative, but we hope that the examples we provide will stimulate discussion about what will advance the field of IBD therapy.
Collapse
Affiliation(s)
- Scott E. Plevy
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Stephan R. Targan
- Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
277
|
Haghikia A, Perrech M, Pula B, Ruhrmann S, Potthoff A, Brockmeyer NH, Goelz S, Wiendl H, Lindå H, Ziemssen T, Baranzini SE, Käll TB, Bengel D, Olsson T, Gold R, Chan A. Functional energetics of CD4+-cellular immunity in monoclonal antibody-associated progressive multifocal leukoencephalopathy in autoimmune disorders. PLoS One 2011; 6:e18506. [PMID: 21533133 PMCID: PMC3080364 DOI: 10.1371/journal.pone.0018506] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Accepted: 03/09/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Progressive multifocal leukoencephalopathy (PML) is an opportunistic central nervous system- (CNS-) infection that typically occurs in a subset of immunocompromised individuals. An increasing incidence of PML has recently been reported in patients receiving monoclonal antibody (mAb) therapy for the treatment of autoimmune diseases, particularly those treated with natalizumab, efalizumab and rituximab. Intracellular CD4(+)-ATP-concentration (iATP) functionally reflects cellular immunocompetence and inversely correlates with risk of infections during immunosuppressive therapy. We investigated whether iATP may assist in individualized risk stratification for opportunistic infections during mAb-treatment. METHODOLOGY/PRINCIPAL FINDINGS iATP in PHA-stimulated, immunoselected CD4(+)-cells was analyzed using an FDA-approved assay. iATP of mAb-associated PML (natalizumab (n = 8), rituximab (n = 2), efalizumab (n = 1)), or other cases of opportunistic CNS-infections (HIV-associated PML (n = 2), spontaneous PML, PML in a psoriasis patient under fumaric acids, natalizumab-associated herpes simplex encephalitis (n = 1 each)) was reduced by 59% (194.5±29 ng/ml, mean±SEM) in comparison to healthy controls (HC, 479.9±19.8 ng/ml, p<0.0001). iATP in 14 of these 16 patients was at or below 3(rd) percentile of healthy controls, similar to HIV-patients (n = 18). In contrast, CD4(+)-cell numbers were reduced in only 7 of 15 patients, for whom cell counts were available. iATP correlated with mitochondrial transmembrane potential (ΔΨ(m)) (iATP/ΔΨ(m)-correlation:tau = 0.49, p = 0.03). Whereas mean iATP of cross-sectionally analysed natalizumab-treated patients was unaltered (448.7±12 ng/ml, n = 150), iATP was moderately decreased (316.2±26.1 ng/ml, p = 0.04) in patients (n = 7) who had been treated already during the pivotal phase III trials and had received natalizumab for more than 6 years. 2/92 (2%) patients with less than 24 months natalizumab treatment revealed very low iATP at or below the 3(rd) percentile of HC, whereas 10/58 (17%) of the patients treated for more than 24 months had such low iATP-concentrations. CONCLUSION Our results suggest that bioenergetic parameters such as iATP may assist in risk stratification under mAb-immunotherapy of autoimmune disorders.
Collapse
Affiliation(s)
- Aiden Haghikia
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Moritz Perrech
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Bartosz Pula
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Sabrina Ruhrmann
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Anja Potthoff
- Department of Dermatology and HIV Competence Center, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Norbert H. Brockmeyer
- Department of Dermatology and HIV Competence Center, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Susan Goelz
- Biogen Idec, Cambridge, Massachusetts, United States of America
| | - Heinz Wiendl
- Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Hans Lindå
- Neurology Unit, Department of Medicine, Karolinska Institute, Danderyd Hospital, Stockholm, Sweden
| | - Tjalf Ziemssen
- Department of Neurology, Technical University, Dresden, Germany
| | - Sergio E. Baranzini
- Department of Neurology at the University of California San Francisco, San Francisco, California, United States of America
| | - Tor-Björn Käll
- Clinic of Internal Medicine Södersjukhuset, Stockholm, Sweden
| | - Dietmar Bengel
- Neurologic Clinic, Oberschwabenklinik, Ravensburg, Germany
| | - Tomas Olsson
- Department of Neurology, Karolinska Hospital, Stockholm, Sweden
| | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
| | - Andrew Chan
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, Bochum, Germany
- * E-mail:
| |
Collapse
|
278
|
Annaloro C, Onida F, Lambertenghi Deliliers G. Autologous hematopoietic stem cell transplantation in autoimmune diseases. Expert Rev Hematol 2011; 2:699-715. [PMID: 21082959 DOI: 10.1586/ehm.09.60] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The term 'autoimmune diseases' encompasses a spectrum of diseases whose clinical manifestations and, possibly, biological features vary widely. The results of conventional treatment are considered unsatisfactory in aggressive forms, with subsets of patients having short life expectancies. Relying on wide experimental evidence and more feeble clinical data, some research groups have used autologous hematopoietic stem cell transplantation (HSCT) in the most disabling autoimmune diseases with the aim of resetting the patient's immune system. Immunoablative conditioning regimens are preferred over their myeloablative counterparts, and some form of in vivo and/or ex vivo T-cell depletion is generally adopted. Despite 15 years' experience, published controlled clinical trials are still lacking, with the evidence so far available coming from pilot studies and registry surveys. In multiple sclerosis, clinical improvement, or at least lasting disease stabilization, can be achieved in the majority of the patients; nevertheless, the worst results are observed in patients with progressive disease, where no benefit can be expected from conventional therapy. Concerning rheumatologic diseases, wide experience has been acquired in systemic sclerosis, with long-term improvements in cutaneous disease being frequently reported, although visceral involvement remains unchanged at best. Autografting has proved to be barely effective in rheumatoid arthritis and quite toxic in juvenile idiopathic arthritis, whereas it leads to clinical remission and the reversal of visceral impairment in the majority of patients with systemic lupus erythematosus. A promising indication is Crohn's disease, in which long-term endoscopic remission is frequently observed. Growing experience with autologous HCST in autoimmune diseases has progressively reduced concerns about transplant-related mortality and secondary myelodysplasia/leukemia. Therefore, a sustained complete remission seems to be within the reach of autografting in some autoimmune diseases; in others, the indications, risks and benefits of autografting need to be better defined. Consequently, the search for new drugs should also be encouraged.
Collapse
Affiliation(s)
- Claudio Annaloro
- Bone Marrow Transplantation Center-Hematology I, Fondazione Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena, University of Milan, Via Francesco Sforza 35, Milan, Italy
| | | | | |
Collapse
|
279
|
Fontoura P. Monoclonal antibody therapy in multiple sclerosis: Paradigm shifts and emerging challenges. MAbs 2011; 2:670-81. [PMID: 21124072 DOI: 10.4161/mabs.2.6.13270] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Therapeutic approaches to multiple sclerosis (MS) are based on altering the functions of the immune system, either by using broad immunosuppressive drugs used for transplantation rejection and rheumatology, or by modulating them more discreetly with beta interferon and synthetic amino-acid copolymers. These strategies are only partially successful, have important safety and tolerability limitations, and have shown to be mostly effective in earlier stages of the disease, in which acute relapses dominate the clinical picture. For progressive phenotypes of MS there are currently no effective therapeutic options. As very specific and potent immunosuppressive agents, monoclonal antibodies (mAbs) may offer considerable advantages over other therapies for MS. During the last decade, anti-a4 integrin natalizumab became the first approved mAb for treatment of relapsing MS, after convincingly demonstrating clinically significant effects on two large Phase 3 trials. Moreover, the concept of disease remission was introduced for the first time, to describe patients that show no signs of clinical or imaging markers of disease activity during therapy with natalizumab. Of the mAbs under development for MS, alemtuzumab and rituximab have also shown promising evidence of effectiveness, and potentially expanded the therapeutic horizon to reversal of disease progression in early relapsing patients, and progressive patients who previously had not been studied. However, the appearance of progressive multifocal leukoencephalopathy (PML) in natalizumab-treated MS patients, as well as in patients with lymphoma, lupus and rheumatoid arthritis treated with rituximab, and autoimmune-type complications in alemtuzumab-treated MS patients underlines the fact that extended efficacy comes with significant clinical risks. The challenge is then how best to utilize therapies that have evidently superior efficacy in a chronic disease of young adults, to obtain the best benefit-risk ratio, and how to monitor and prevent emergent safety concerns.
Collapse
Affiliation(s)
- Paulo Fontoura
- Roche Pharmaceuticals, Pharma Research and Exploratory Development, Translational Medicine CNS, Basel, Switzerland.
| |
Collapse
|
280
|
Krieger S. Multiple Sclerosis Therapeutic Pipeline: Opportunities and Challenges. ACTA ACUST UNITED AC 2011; 78:192-206. [DOI: 10.1002/msj.20241] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
281
|
Keyser FD. Choice of Biologic Therapy for Patients with Rheumatoid Arthritis: The Infection Perspective. Curr Rheumatol Rev 2011; 7:77-87. [PMID: 22081766 PMCID: PMC3182090 DOI: 10.2174/157339711794474620] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2010] [Revised: 07/30/2010] [Accepted: 11/10/2010] [Indexed: 12/27/2022]
Abstract
Biologicals revolutionized the treatment of Rheumatoid Arthritis (RA). The targeted suppression of key inflammatory pathways involved in joint inflammation and destruction allows better disease control, which, however, comes at the price of an elevated infection risk due to relative immunosuppression. The disease-related infection risk and the infection risk associated with the use of TNF-α inhibitors (infliximab, adalimumab, etanercept, golimumab and certolizumab pegol), rituximab, abatacept and tocilizumab are discussed. Risk factors clinicians need to take into account when selecting the most appropriate biologic therapy for RA patients, as well as precautions and screening concerning a number of specific infections, such as tuberculosis, intracellular bacterial infections, reactivation of chronic viral infections and HIV are reviewed.
Collapse
|
282
|
Infectious Complications Associated with Immunomodulating Biologic Agents. Hematol Oncol Clin North Am 2011; 25:117-38. [DOI: 10.1016/j.hoc.2010.11.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
283
|
Regulation of human neurotropic JC virus replication by alternative splicing factor SF2/ASF in glial cells. PLoS One 2011; 6:e14630. [PMID: 21297941 PMCID: PMC3031499 DOI: 10.1371/journal.pone.0014630] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2010] [Accepted: 01/09/2011] [Indexed: 11/19/2022] Open
Abstract
Background The human neurotropic virus, JC virus (JCV), is the etiologic agent of the fatal demyelinating disease of the central nervous system, Progressive Multifocal Leukoencephlopathy (PML) that is seen primarily in immunodeficient individuals. Productive infection of JCV occurs only in glial cells, and this restriction is, to a great extent, due to the activation of the viral promoter that has cell type-specific characteristics. Earlier studies led to the hypothesis that glial-specific activation of the JCV promoter is mediated through positive and negative transcription factors that control reactivation of the JCV genome under normal physiological conditions and suppress its activation in non-glial cells. Methodololgy/Principal Findings Using a variety of virological and molecular biological approaches, we demonstrate that the alternative splicing factor SF2/ASF has the capacity to exert a negative effect on transcription of the JCV promoter in glial cells through direct association with a specific DNA sequence within the viral enhancer/promoter region. Our results show that down-regulation of SF2/ASF in fetal and adult glial cells increases the level of JCV gene expression and its replication indicating that negative regulation of the JCV promoter by SF2/ASF may control reactivation of JCV replication in brain. Conclusions/Significance Our results establish a new regulatory role for SF2/ASF in controlling gene expression at the transcriptional level.
Collapse
|
284
|
Abstract
There is a need for methods to improve the diagnosis, patient staging and evaluation of therapeutic response in patients with autoimmune conditions to improve patient care. Inflammatory bowel disease (IBD) and rheumatoid arthritis (RA) are two inflammatory diseases characterized by involvement of innate and adaptive immune components that change the metabolic state of their respective target tissues, thus providing an opportunity for molecular imaging probes to detect such changes. Optimally, such probes and the imaging methods employed would be non-invasive, robust and reproducible, give a quantitative result, report on the status of the affected tissue(s) and respond to the effects of a therapeutic molecule. Positron emission tomography (PET) and single photon emission computed tomography (SPECT) are nuclear imaging approaches that have the potential to satisfy such requirements. In this review, the work to date and the potential of PET and SPECT imaging probes in these two inflammatory conditions, IBD and RA, are discussed.
Collapse
Affiliation(s)
- Helen J McBride
- Inflammation Research, Amgen, Inc., One Amgen Center Drive, MS: 29-1-B, Thousand Oaks, CA 91320, USA.
| |
Collapse
|
285
|
Khamashta MA, Ramos-Casals M. Life-Threatening Complications of Biological Therapies. Autoimmune Dis 2011. [PMCID: PMC7120365 DOI: 10.1007/978-0-85729-358-9_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
The last decade was characterized by the successive introduction of several biological agents for the treatment of autoimmune rheumatic diseases (ARD). Randomized controlled trials (RCT) proved them to have globally acceptable safety and tolerability profiles. However, life-threatening complications are rare events and RCT are underpowered to detect them. As these drugs became more widely prescribed in clinical practice, and particularly, having the information from multiple national biologics registries available, serious adverse events became perceptible. Infection remains the major concern, but other serious and life-threatening complications have emerged, such as malignancies, congestive heart failure, demyelinating disorders, and drug-induced autoimmune syndromes. Several of these are correlated with either the underlying disease or concomitant immunosuppressive medication. Most of them can be avoided by the adoption of preventive measures and an early proper management might significantly change the outcome. Awareness of the possible serious side effects is of utmost importance for a safer use of biological agents. In this chapter, we aim to describe the most commonly reported life-threatening complications of biological therapies in the literature – including those with antitumor necrosis factor agents, rituximab, abatacept, tocilizumab, and anakinra. Risk groups are identified and strategies for the prevention and initial management are included.
Collapse
Affiliation(s)
- Munther A. Khamashta
- Rayne Institute, Lupus Research Unit, St. Thomas' Hospital, London, SE1 7EH United Kingdom
| | - Manuel Ramos-Casals
- Barcelona, Surgey, Hospital Clinic, Calle Villarroel, 170, Barcelona, 08036 Spain
| |
Collapse
|
286
|
Horga A, Tintoré M. Natalizumab for relapsing-remitting multiple sclerosis. NEUROLOGÍA (ENGLISH EDITION) 2011. [DOI: 10.1016/s2173-5808(11)70082-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
287
|
Chiarchiaro J, McLendon RE, Buckley PJ, Laskowitz DT. Progressive Multifocal Leukoencephalopathy With Occult Waldenström Macroglobulinemia. J Clin Oncol 2010; 28:e759-61. [DOI: 10.1200/jco.2010.30.7447] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
288
|
Abstract
Drug-induced adverse reactions represent a significant health problem in developed countries. These events cause 5% of hospital admissions and are one of the main causes of mortality. Neurological manifestations are among the most frequent. This article reviews catastrophic cerebrovascular situations and confusional syndromes, as well as epilepsy, structural encephalopathy, neuromuscular disorders, catastrophic movement disorders and infections, all of which can be drug-induced.
Collapse
|
289
|
Meyer-Olson D, Hoeper K, Schmidt RE. [Infectious complications of biologic therapy in patients with rheumatoid arthritis]. Z Rheumatol 2010; 69:879-88. [PMID: 21128049 DOI: 10.1007/s00393-010-0677-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The introduction of biological disease-modifying drugs (DMARDs) has substantially improved the treatment options for patients with rheumatoid arthritis. However, infectious complications represent the most common side effects of these drugs, including severe infections as well as rare opportunistic infections. Treating patients on biological DMARDs is therefore one of the biggest challenges in rheumatology care. The present review describes the current state of knowledge regarding frequency and type of infectious complications associated with biological DMARDs. The article focuses mainly on risk management, in particular on diagnosis and recurrence prevention of tuberculosis and reactivation of hepatitis B virus infection. Furthermore, we discuss the importance of vaccinations in primary disease prevention in patients with rheumatoid arthritis.
Collapse
Affiliation(s)
- D Meyer-Olson
- Klinik für Immunologie und Rheumatologie, Medizinische Hochschule Hannover, Carl-Neuberg-Strasse 1, Hannover, Germany.
| | | | | |
Collapse
|
290
|
|
291
|
Abstract
Psoriasis is one of the commonest chronic inflammatory disorders. Its cause is unknown, but a wealth of studies indicate that the disease results from a complex and dynamic interplay between genetic and environmental factors that trigger an excessive inflammatory response in the skin. Dendritic cells and effector T-cells are central in the development of the psoriastic lesion, and cytokines produced by these cells stimulate keratinocytes to proliferate and increase the migration of inflammatory cells into the skin, promoting epidermal hyperplasia and inflammation. Understanding the immunology of the psoriatic plaque has led to new therapeutic options and novel candidates for immunomodulation, and has changed the ways psoriatic patients are managed.
Collapse
|
292
|
Weger W. Current status and new developments in the treatment of psoriasis and psoriatic arthritis with biological agents. Br J Pharmacol 2010; 160:810-20. [PMID: 20590580 DOI: 10.1111/j.1476-5381.2010.00702.x] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Psoriasis is a chronic inflammatory disease affecting 1-3% of the general population. Among psoriatic patients, 5-40% are affected by psoriatic arthritis. Due to the chronic nature of the disease, patients suffer from substantial psychological and financial burdens, thus adding to a significantly impaired quality of life. Traditional systemic therapies for psoriasis, such as methotrexate, cyclosporin A, retinoids or PUVA therapy, have a potential for long-term toxicity and may not always provide sufficient improvement of the disease. The development of novel therapies targeting key steps in the pathogenesis of psoriasis and psoriatic arthritis now provide new and efficient treatment options. Biological therapies for the treatment of psoriasis and/or psoriatic arthritis are defined by their mode of action and can be classified into three categories: the T-cell modulating agents (alefacept and efalizumab), the inhibitors of tumour necrosis factor-alpha (TNFalpha blockers, e.g. adalimumab, certolizumab, etanercept, golimumab and infliximab) and the inhibitors of interleukin (IL) 12 and IL-23 (e.g. ustekinumab and briakinumab). This article provides a brief overview of the currently approved biological agents in the European Union and of some newer agents, such as briakinumab, certolizumab and golimumab.
Collapse
Affiliation(s)
- Wolfgang Weger
- Department of Dermatology, Medical University of Graz, Graz, Austria.
| |
Collapse
|
293
|
Tuccori M, Focosi D, Blandizzi C, Pelosini M, Montagnani S, Maggi F, Pistello M, Antonioli L, Fornai M, Pepe P, Rossi G, Petrini M. Inclusion of rituximab in treatment protocols for non-Hodgkin's lymphomas and risk for progressive multifocal leukoencephalopathy. Oncologist 2010; 15:1214-9. [PMID: 21041380 DOI: 10.1634/theoncologist.2010-0098] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Objectives. Rituximab is an anti-CD20 monoclonal antibody that promotes better treatment outcomes in patients with non-Hodgkin's lymphoma (NHL). Case series of progressive multifocal leukoencephalopathy (PML) in patients receiving rituximab within polychemotherapy regimens have led to the introduction of a black box warning, but no risk estimation has ever been provided. Methods. We performed a retrospective, monocentric cohort study on 976 NHL patients diagnosed in 1994-2008, including 517 patients who received at least one dose of rituximab. Results. Inclusion of rituximab into standard chemotherapy regimens for NHL caused a significantly higher incidence of PML cases (rate difference, 2.2 every 1,000 patient-years; 95% confidence interval, 0.1-4.3). Interpretation. Based on this finding, clinical surveillance of PML-related symptoms is recommended in NHL patients exposed to rituximab.
Collapse
Affiliation(s)
- Marco Tuccori
- Department of Experimental Pathology, University of Pisa, Pisa, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
294
|
Maródi L, Casanova JL. Primary immunodeficiencies may reveal potential infectious diseases associated with immune-targeting mAb treatments. J Allergy Clin Immunol 2010; 126:910-7. [DOI: 10.1016/j.jaci.2010.08.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2010] [Revised: 07/16/2010] [Accepted: 08/06/2010] [Indexed: 01/13/2023]
|
295
|
Kumar D, Bouldin TW, Berger RG. A case of progressive multifocal leukoencephalopathy in a patient treated with infliximab. ARTHRITIS AND RHEUMATISM 2010; 62:3191-3195. [PMID: 20722036 DOI: 10.1002/art.27687] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
We describe a 72-year-old white man with erosive rheumatoid arthritis in whom subacute neurologic and psychiatric symptoms developed after 3 years of treatment with infliximab, prednisone, and methotrexate. White matter demyelination was seen on magnetic resonance imaging of the brain, and progressive multifocal leukoencephalopathy (PML) was ultimately confirmed by brain biopsy. The patient was treated with supportive therapy and discontinuation of disease-modifying antirheumatic drugs, resulting in stabilization of the disease process. The patient survived, but neurologic and cognitive deficits persisted. The distribution and pathology of this patient's disease are unique from almost all reported incidents of oral methotrexate-associated leukoencephalopathy. The pathogenesis of disease may be linked to a T cell-mediated process that is potentially impacted by infliximab. This case provides the first reported evidence that PML can be seen in association with infliximab therapy.
Collapse
Affiliation(s)
- Deepak Kumar
- University of North Carolina at Chapel Hill, Chapel Hill, NC 27517, USA
| | | | | |
Collapse
|
296
|
Harrington EP, Zhao C, Fancy SP, Kaing S, Franklin RJ, Rowitch DH. Oligodendrocyte PTEN is required for myelin and axonal integrity, not remyelination. Ann Neurol 2010; 68:703-16. [PMID: 20853437 PMCID: PMC2966537 DOI: 10.1002/ana.22090] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Repair of myelin injury in multiple sclerosis may fail, resulting in chronic demyelination, axonal loss, and disease progression. As cellular pathways regulated by phosphatase and tensin homologue deleted on chromosome 10 (PTEN; eg, phosphatidylinositol-3-kinase [PI-3K]) have been reported to enhance axon regeneration and oligodendrocyte maturation, we investigated potentially beneficial effects of Pten loss of function in the oligodendrocyte lineage on remyelination. METHODS We characterized oligodendrocyte numbers and myelin sheath thickness in mice with conditional inactivation of Pten in oligodendrocytes, Olig2-cre, Pten(fl/fl) mice. Using a model of central nervous system demyelination, lysolecithin injection into the spinal cord white matter, we performed short- and long-term lesioning experiments and quantified oligodendrocyte maturation and myelin sheath thickness in remyelinating lesions. RESULTS During development, we observed dramatic hypermyelination in the corpus callosum and spinal cord. Following white matter injury, however, there was no detectable improvement in remyelination. Moreover, we observed progressive myelin sheath abnormalities and massive axon degeneration in the fasciculus gracilis of mutant animals, as indicated by ultrastructure and expression of SMI-32, amyloid precursor protein, and caspase 6. INTERPRETATION These studies indicate adverse effects of chronic Pten inactivation (and by extension, activation PI-3K signaling) on myelinating oligodendrocytes and their axonal targets. We conclude that PTEN function in oligodendrocytes is required to regulate myelin thickness and preserve axon integrity. In contrast, PTEN is dispensable during myelin repair, and its inactivation confers no detectable benefit.
Collapse
Affiliation(s)
- Emily P. Harrington
- Departments of Pediatrics and Neurosurgery, Eli and Edyth Broad Institute for Stem Cell Research and Regeneration Medicine and Howard Hughes Medical Institute, San Francisco, CA, 94143, USA
- Medical Scientist Training Program, University of California San Francisco, 513 Parnassus Avenue, San Francisco, CA, 94143, USA
| | - Chao Zhao
- MRC Centre for Stem Cell Biology and Regenerative Medicine and Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK
| | - Stephen P.J. Fancy
- Departments of Pediatrics and Neurosurgery, Eli and Edyth Broad Institute for Stem Cell Research and Regeneration Medicine and Howard Hughes Medical Institute, San Francisco, CA, 94143, USA
| | - Sovann Kaing
- Departments of Pediatrics and Neurosurgery, Eli and Edyth Broad Institute for Stem Cell Research and Regeneration Medicine and Howard Hughes Medical Institute, San Francisco, CA, 94143, USA
| | - Robin J.M. Franklin
- MRC Centre for Stem Cell Biology and Regenerative Medicine and Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK
| | - David H. Rowitch
- Departments of Pediatrics and Neurosurgery, Eli and Edyth Broad Institute for Stem Cell Research and Regeneration Medicine and Howard Hughes Medical Institute, San Francisco, CA, 94143, USA
| |
Collapse
|
297
|
Dahan A, Amidon GL, Zimmermann EM. Drug targeting strategies for the treatment of inflammatory bowel disease: a mechanistic update. Expert Rev Clin Immunol 2010; 6:543-50. [PMID: 20594127 DOI: 10.1586/eci.10.30] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The therapeutic management of inflammatory bowel disease (IBD) represents the perfect scenario for drug targeting to the site(s) of action. While existing formulation-based targeting strategies include rectal dosage forms and oral systems that target the colon by pH-, time-, microflora- and pressure-triggered drug release, novel approaches for site-specific delivery in IBD therapy will target the inflamed intestine per se rather than intestinal region. The purpose of this article is to present a mechanistic update on the strategies employed to achieve minimal systemic exposure accompanied by maximal drug levels in the inflamed intestinal tissue. The introduction of biological agents, micro/nanoparticulate carriers including liposomes, transgenic bacteria, and gene therapy opportunities are discussed, as well as the challenges remaining to be achieved in the targeted treatment of IBD.
Collapse
Affiliation(s)
- Arik Dahan
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| | | | | |
Collapse
|
298
|
Chaudhry V, Cornblath DR. An open-label trial of rituximab (Rituxan®) in multifocal motor neuropathy. J Peripher Nerv Syst 2010; 15:196-201. [DOI: 10.1111/j.1529-8027.2010.00270.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
299
|
[Therapeutic monoclonal antibodies in clinical neurology]. DER NERVENARZT 2010; 81:753-64; quiz 765-6. [PMID: 20232033 DOI: 10.1007/s00115-010-2947-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Recombinant monoclonal antibodies break new ground in the treatment of immune-mediated nerve and muscle disorders but also of neurodegenerative diseases, in the field of neuro-oncology and in pain therapy, as they allow molecular targeting of defined cell populations or key pathophysiological molecules. However, safety risks might accompany a high efficacy. Basic understanding of this increasingly important class of agents and a steady update of knowledge, in particular on safety aspects, are therefore key requirements for responsible use based on an individual benefit-risk assessment.
Collapse
|
300
|
Neu U, Maginnis MS, Palma AS, Ströh LJ, Nelson CDS, Feizi T, Atwood WJ, Stehle T. Structure-function analysis of the human JC polyomavirus establishes the LSTc pentasaccharide as a functional receptor motif. Cell Host Microbe 2010; 8:309-19. [PMID: 20951965 PMCID: PMC2957469 DOI: 10.1016/j.chom.2010.09.004] [Citation(s) in RCA: 154] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Revised: 07/16/2010] [Accepted: 08/17/2010] [Indexed: 11/28/2022]
Abstract
The human JC polyomavirus (JCV) causes a fatal demyelinating disease, progressive multifocal leukoencephalopathy (PML), in immunocompromised individuals. Current treatment options for PML are inadequate. Sialylated oligosaccharides and the serotonin receptor are known to be necessary for JCV entry, but the molecular interactions underlying JCV attachment remain unknown. Using glycan array screening and viral infectivity assays, we identify a linear sialylated pentasaccharide with the sequence NeuNAc-α2,6-Gal-β1,4-GlcNAc-β1,3-Gal-β1,4-Glc (LSTc) present on host glycoproteins and glycolipids as a specific JCV recognition motif. The crystal structure of the JCV capsid protein VP1 was solved alone and in complex with LSTc. It reveals extensive interactions with the terminal sialic acid of the LSTc motif and specific recognition of an extended conformation of LSTc. Mutations in the JCV oligosaccharide-binding sites abolish cell attachment, viral spread, and infectivity, further validating the importance of this interaction. Our findings provide a powerful platform for the development of antiviral compounds.
Collapse
Affiliation(s)
- Ursula Neu
- Interfaculty Institute for Biochemistry, University of Tübingen, D-72076 Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|