Structural formula of isoflurane
Find information on thousands of medical conditions and prescription drugs.

Isoflurane


Isoflurane (1-chloro-2,2,2-trifluoroethyl difluoromethyl ether) is a halogenated ether used for inhalation anesthesia. Together with enflurane and halothane it replaced the flammable ethers used in the pioneer days of surgery. Its use in human medicine is now starting to decline, being replaced with sevoflurane, desflurane and the intravenous anaesthetic propofol. Isoflurane is still frequently used for veterinary anaesthesia. more...

Home
Diseases
Medicines
A
B
C
D
E
F
G
H
I
Ibuprofen
Idarubicin
Idebenone
IFEX
Iloprost
Imatinib mesylate
Imdur
Imipenem
Imipramine
Imiquimod
Imitrex
Imodium
Indahexal
Indapamide
Inderal
Indocin
Indometacin
Infliximab
INH
Inosine
Intal
Interferon gamma
Intralipid
Invanz
Invirase
Iontocaine
Iotrolan
Ipratropium bromide
Iproniazid
Irbesartan
Iressa
Irinotecan
Isocarboxazid
Isoflurane
Isohexal
Isoleucine
Isomonit
Isoniazid
Isoprenaline
Isordil
Isosorbide
Isosorbide dinitrate
Isosorbide mononitrate
Isotretinoin
Itraconazole
Ivermectin
J
K
L
M
N
O
P
Q
R
S
T
U
V
W
X
Y
Z

Isoflurane is always administered in conjunction with air and/or pure oxygen. Often nitrous oxide is also used. Although its physical properties means that anaesthesia can be induced more rapidly than with halothane, its pungency can irritate the respiratory system, negating this theoretical advantages conferred by its physical properties. It is usually used to maintain a state of general anesthesia that has been induced with another drug, such as thiopentone or propofol.

A major advantage of isoflurane is that it is no longer patented, and hence very economical to use.

It vaporizes readily, but is a liquid at room temperature. It is completely non-flammable.

Physical properties

Read more at Wikipedia.org


[List your site here Free!]


Intravenous immunoglobulin ameliorates experimental autoimmune encephalomyelitis and reduces neuropathological abnormalities when administered prophylactically
From Neurological Research, 9/1/05 by Jorgensen, Signe Humle

Background and methods: Immunomodulation with intravenous immunoglobulin (IVIG) represents a way of interfering with the disease process in multiple sclerosis (MS). In this study, the effects of IVIG on neurological symptoms and central nervous system (CNS) pathology were evaluated in experimental autoimmune encephalomyelitis (EAE), an MS animal model. EAE was induced in susceptible Dark Agouti rats by active immunization with a spinal cord homogenate, and infusions of 1 g/kg IVIG were given prophy/actically or therapeutically.

Results: The administration of IVIG at the time of immunization significantly suppressed the development of neurological symptoms compared with infusions of placebo (mean EAE score 0.6±0.3 versus 2.3±0.4). Moreover, the prophylactic IVIG administration resulted in a significant inhibition of the inflammatory response in CNS tissue (inflammation score 1.1±0.2 versus 1.8±0.2 after placebo). No beneficial effects were obtained by therapeutic IVIG infusions as the EAE disease course and the degree of inflammation and demyelination in the CNS were not different from animals receiving treatment with placebo.

Conclusions: The present study indicates that IVIG reduces the symptoms of EAE by suppression of the CNS inflammation that characterizes CNS pathology in these animals. Taking into account data from clinical trials of IVIG in MS, the results further suggest that IVIG acts primarily during the induction phase of the immune response thus preventing the development of relapses in MS. [Neural Res 2005; 27: 591-597]

Keywords: Intravenous immunoglobulin; immunoglobulin G; experimental autoimmune encephalomyelitis; multiple sclerosis; Dark Agouti rat

INTRODUCTION

Multiple sclerosis (MS) is an autoimmune disease of the central nervous system. The pathology of MS is characterized by white matter lesions of focal demyelination and axonal damage in the brain and spinal cord1,2. Initially, most patients have a relapsingremitting disease course that typically shifts towards ongoing progression later, i.e. secondary progressive MS1. The cause of MS still has to be resolved, but the interaction of genetic predisposition and yet unidentified environmental factors are thought to be responsible3,4. Because of the lack of measurable trigger factors, the most important diagnostic criterion is the demonstration of dissemination of demyelinated central nervous system (CNS) lesions in both time and space, generally achieved by magnetic resonance imaging5. Pathologically, the hallmark of MS is the destruction of myelin and axonal degeneration driven by both T lymphocytes and antibodies2,6,7. Current therapies are only partially effective in attenuating disease activity, and MS is, therefore, still a disease with unmet medical needs.

Clinical trials have shown that immunoglobulin for intravenous administration (IVIG) has the potential to reduce the disease activity in MS8-10. However, the mechanisms by which IVIG may interfere with the pathophysiology of MS are not yet fully understood. The effects of IVIG treatment presumably owe to the variety of antibody specificities present in the IVIG preparations. IVIG may target harmful autoantibodies by anti-idiotypic interactions11,12, but may also inhibit inflammatory cells by binding through the constant Fc region of the immunoglobulin molecule13,14. Furthermore, IVIG may inhibit damage mediated by the complement system15,16 and may be involved in myelin repair through the process of remyelination17,18.

The classical animal model for studying MS, experimental autoimmune encephalomyelitis (EAE), is an inflammatory disease of the CNS primarily mediated by T cells19. EAE with a protracted and relapsing disease course resembling MS can be induced by the immunization of susceptible rat strains with CNS antigens20,21. Like MS, EAE in rodents may also be associated with axonal damage and neurodegeneration22,23. IVIG administration has previously been reported to significantly ameliorate the neurological symptoms of EAE24,25. In the present study, we induced EAE in the susceptible rat strain Dark Agouti (DA) in order to study the effects of IVIG treatment on the pathological changes in the CNS. The animals were treated with infusions of IVIG administered prophylactically or therapeutical Iy, and the CNS histopathology was studied during the acute attack at day 11 and 4 weeks after the induction of EAE.

MATERIALS AND METHODS

Animals

A protracted and relapsing type of EAE was induced in male rats of the susceptible inbred strain DA (180-200 g; Harlan, Horst, The Netherlands). The animals were acclimatized for a minimum of 5 days and maintained on a 12-hour light-dark cycle. Food and water were given ad libitum. The experiments were conducted according to the guidelines of the Danish Committee for Animal Experiments.

Induction of EAE

EAE was induced by subcutaneous inoculation of 0.2 ml encephalitogenic emulsion at the base of the tail. The emulsion was prepared as follows: spinal cord tissue obtained from normal male DA rats (>225 g) was homogenized in saline (1 g tissue/1 ml 0.9% NaCl, pH 7.4) and emulsified in an equal volume of incomplete Freund's adjuvant (IFA) (Difco Laboratories Inc., Detroit, Ml, USA). During the immunization, the animals were kept under light anaesthesia by isoflurane inhalation (Abbott Scandinavia AB, Solna, Sweden). The control rats were inoculated with saline.

Evaluation of EAE symptoms

Throughout the study, the animals were evaluated on a daily basis for weight loss and neurological symptoms of active disease. The EAE symptoms were assessed according to the following scoring system: 0, no clinical signs; 1, tail paralysis; 2, mild-to-moderate hind limb paresis; 3, severe hind limb paresis; 4, paralysis of the limbs; 5, tetraplegia; 6, moribund state or death from EAE.

IVIG administration

Human IVIG (10% IgG; Bayer AG, Leverkusen, Germany) was administered on 2 consecutive days: as prophylactic treatment at the time of EAE induction at day 0 and 1 post-immunization (p.L), or in a therapeutic treatment protocol at the beginning of the acute EAE attack at days 8 and 9 p.i. (EAE+ IVIG, n=12 per group). A dose of 1 g IgG/kg was administered into a tail vein at an infusion rate of 0.2 ml/minute. The control animals received an intravenous infusion of either placebo solution (0.1% albumin in 10% maltose; Bayer AG, Leverkusen, Germany) (EAE+ placebo, n=12 per group) or saline (0.9% NaCI, pH 7.4) (EAE control, n=7; saline control, n=7).

Histological evaluation

The experiments were stopped at two different time points: during the acute EAE attack at day 11 p.i., or during the remission of the disease at day 28 p.i.. The animals were anaesthetized with isoflurane and perfused intracardially with phosphate-buffered saline. The brain and spinal cord segments C^sub 5-7^ and L^sub 1-3^ were dissected and fixed in 4% paraformaldehyde. The tissues were then dehydrated, embedded in paraffin and 3-µm sections were cut using a microtome. Random numbers were assigned to the sections, and the study remained blinded until after the final evaluation. The sections were rehydrated through xylene and ethanol before staining with haematoxylin and eosin or Kluver-Barrera stain for the assessment of inflammation and demyelination. The degree of inflammatory changes in the CNS was evaluated according to the following scoring system: grade 0, no inflammation; grade 1, minor perivascular infiltration; grade 2, several perivascular and parenchymal infiltrations; grade 3, maximal inflammatory response and demyelination.

Statistical analysis

Data are presented as mean±SEM. The Student's t-test was used for the comparison of changes in body weight. Clinical and histological scores were compared for significant differences between the treatment groups by the Mann-Whitney rank sum test. The Spearman rank order correlation was used to determine the association between the clinical or histological scoring and body weight changes. Probability values below 0.05 were considered statistically significant.

RESULTS

Effects of prophylactic IVIG on development of the acute EAE attack

In the saline control group the animals resumed normal growth curves a few days after the inoculation and infusion procedures. The animals in this group did not develop any sign of disease. The rats injected with the encephalitogenic emulsion receiving no further treatment (EAE controls and EAE+placebo group) developed an acute EAE attack at days 6-7 p.i. The initial neurological symptoms included loss of tail tone and were accompanied by a reduction in body weight (Figures 1 and 2). The observed reduction in body weight was aggravated until the acute EAE attack reached its maximum at days 10-11 p.i. At this time point, the EAE symptoms also included paralysis of the tail and paresis or paralysis of the hind limbs. Intravenous administration of 1 g/kg IVIG at days O and 1 p.i. significantly reduced the loss of body weight during the acute attack (p

Effects of prophylactic versus therapeutic IVIG treatment on long-term EAE

Infusions of IVIG were administered either prophylactically at the time of EAE induction at days 0 and 1 p.i. or therapeutically at the beginning of the acute EAE attack at days 8 and 9 p.i. After immunization, the animals were monitored for 28 days. The symptoms of EAE were observed 1 week after the immunization, where the rats in the placebo group had a profound weight loss. If the animals received the IVIG at days 0-1 p.i., the loss of body weight was significantly reduced (Figure 3A). When IVIC was administered therapeutical Iy at days 8-9 p.i., when the symptoms of EAE were evident, the treatment had no effect on the body weight (Figure 3B). The development of the neurological EAE symptoms coincided with the body weight changes, and prophylactic treatment with immunoglobulin resulted in a less severe course of the disease with only mild symptoms of EAE (Figure 4A). When the infusions of IVIG were given therapeutically during established EAE, no significant treatment effects could be observed (Figure 4B). The EAE incidence after prophylactic IVIG treatment was 75 (9/12) versus 100% (12/12) in animals receiving therapeutic IVIG infusions at days 8-9 p.i.

Histological findings after IVIG treatment

Tissue samples from the CNS were obtained at day 11 p.i. during the acute EAE attack and at day 28 p.i. The principal histopathological findings were a pronounced inflammatory response in both the brain and spinal cord, predominantly as perivascular infiltrations, but demyelination was also observed (Figure 5). Prophylactic administration of IVIG at days 0-1 significantly inhibited the inflammatory response as evaluated by the blinded scoring of the histological findings (Figure 6). The effects of prophylactic IVIG treatment were evident during the acute attack at day 11 p.i. (IVIG average score 1.1±0.2 versus placebo 1.8±0.2, P=0.040) and also at the later stages of EAE at day 28 p.i. (IVIG 1.0±0.1 versus placebo 1.6±0.2, P=0.029). The therapeutic administration of IVIG at days 8-9 p.i. did not affect inflammation or demyelination in the CNS (IVIG 1.6±0.3 versus placebo 1.5±0.2). All the tissue samples from animals immunized with encephalitogenic emulsion exhibited varying degrees of inflammation, whereas all the rats in the saline control group received the inflammation score 0.

Correlation of EAE symptoms and histological findings

Correlation was calculated between the body weight, clinical score at the last day of experiments, and the histopathological scoring. In this study, 86 animals were included of which 76 survived until the end of the experiment at days 11 or 28 p.i.. EAE scores and inflammation scores were positively correlated with a correlation coefficient (r) of 0.72 (P

DISCUSSION

In the present study we found significant effects of IVIG on the acute EAE attack and the long-term disease course. Eoss of body weight and neurological EAE symptoms were only inhibited when IVIG treatment was administered prophylactically at the time of immunization. The therapeutic infusions of IVIG during established EAE did not ameliorate the EAE symptoms and had no beneficial effects on body weight. These observations support previous findings by others24,25. Moreover, we studied the inflammatory response in the brain and spinal cord by histopathology at days 11 and 28 after the induction of EAE. The development of EAE was associated with extensive inflammation in the CNS, and areas of demyelination were also observed. Infusions of IVIG significantly inhibited the histopathology, but only when administered prophylactically. Considering the short duration of treatment applied in this study (days 0 and 1 p.i.), the observations demonstrate that high-dose IVIG treatment is a very effective anti-inflammatory remedy with substantial effects on the CNS lesion formation in EAE. The results also indicate that IVIG acts during the early phase of the immune response in EAE, as no improvement was observed when the infusions were given at days 8-9 p.i. when the symptoms of EAE were evident.

The exact mechanism by which IVIG influences the immune response in CNS inflammation is not known. Considering the many anti-inflammatory and immunomodulatory effects of the polyclonal IVIG preparations, it is likely that several mechanisms act in concert to prevent or ameliorate the development of EAE symptoms. IVIG may be of benefit in autoimmune diseases due to the suppression of harmful autoantibodies through anti-idiotypic interactions12,26, but may also be effective in autoimmune diseases primarily mediated by pathogenic T lymphocytes through inhibitory effects on these cells27,28. The observed efficiency of prophylactic IVIG infusions in EAE is in accordance with the ability of IVIG to inhibit the activation of T cells, which are pivotal in establishing EAE. However, the later stages of the autoimmune T-cell response in EAE are possibly inhibited by the IVIG treatment as well, as it has been shown that EAE by adoptive transfer is inhibited only by IVIG pre-treatment of the activated, encephalitogenic T cells in vitro, not by in vivo IVIG treatment25. In addition to the inhibitory effects on antibodies and lymphocyte function, IVIG may also suppress injury mediated by the complement system15, interfere with the cytokine network29, and affect the function of phagocytes13. These effects of IVIG may contribute to the reduced inflammatory response and demyelination in the CNS, as all of these cellular and humoral immune reactions participate in the development of the EAE disorder20,21. In accordance with the classification of demyelinating CNS lesions described by Lassmann and co-workers2, IVIG may be particularly effective in the type II lesions characterized by the deposition of complement components and antibodies.

Although the modes of actions mentioned above are anti-inflammatory, IVIG treatment may be a suitable strategy in suppressing neurodegeneration. In MS, the pathophysiological sequence of events is thought to involve a primary inflammatory process with cellular infiltration of the CNS followed by demyelination that allows subsequent transection of demyelinated axons in the affected regions1. Apparently, axonal damage is responsible for the irreversible neurological deficits in MS patients and seems to be prevented by protective remyelination in shadow plaques22. In the Theilers murine encephalomyelitis virus (TMEV) model of immune-mediated demyelination it has been shown that immunoglobulin has the potential to act through myelin repair mechanisms30,31. However, the possible remyelinating effects of IVIG in MS have not been confirmed in clinical trials that were designed to evaluate the persistence of stable deficits after optic neuritis or permanent motor deficits in MS32,33. In a recent trial, MS patients receiving IVIG in combination with i.v. methylprednisolone did not recover faster or more completely from acute relapses than patients treated with methylprednisolone alone, and the trial could not corroborate the hypothesis that IVIG enhances remyelination in the acute MS plaque34. IVIG treatment of relapsing-remitting MS has been studied in several controlled trials that demonstrated a significant reduction in the annual relapse rate, a decreased disability score (EDSS), and a reduction in the number of new lesions by MRI after IVIG treatment8,9,35,36. A meta-analysis of four randomized, placebo-controlled studies was recently published confirming that patients with the relapsing type of MS benefit significantly from IVIG treatment10. On the other hand, clinical trials using IVIG infusions for treatment of secondary progressive MS have proven negative37.

The existing experimental and clinical data may seem conflicting as the beneficial effect of IVIG on the relapse rate in MS is high, the magnitude being comparable with that of the first-line compounds interferon-β and glatiramer acetate. The development of EAE symptoms in the experimental model may, however, be considered parallel to a single relapse in the relapsing-remitting type of MS. Therefore, the advantageous effects of IVIG may be regarded as the result of an ability to prevent the development of relapses through various immunomodulatory mechanisms.

To summarize, we observed that IVIG protected against actively induced EAE, and the treatment effect was associated with a significant suppression of inflammatory lesions within the CNS. The data suggest that IVIG acts primarily through the early phase of the immune response, as the therapeutic administration of IVIG had no effect on the course of EAE. It remains to be clarified, which of the numerous anti-inflammatory effects of IVIG are of special importance in CNS inflammation and demyelination.

ACKNOWLEDGEMENTS

The authors acknowledge the technical assistance of Inge Møller, Neurobiology Research Unit, Copenhagen University Hospital, Denmark, Ann Meisler, Jan Lauritzen, and Bodil Sneskov, Laboratory of Neuropathology, Copenhagen University Hospital, Denmark. Parts of this study were presented at the Charcot Symposium 2003, Lisbon, Portugal. Bayer AC, Leverkusen, Germany, provided the intravenous immunoglobulin used in this study. This study was supported by funds from The H0rslev Foundation, Danielsens Foundation, The Danish MS Society, The Dir. Ejnar Johnsen Foundation, Warwara Larsen Foundation, The Augustinus Foundation, Novo Nordisk Foundation, Sigurd and Addie Abrahamson's Legacy, and Karen A. Tolstrups Foundation.

REFERENCES

1 Compston A, Coles A. Multiple sclerosis. Lancet 2002; 359: 1221-1231

2 Lucchinetti C, Bruck W, Parisi J, et al. Heterogeneity of multiple sclerosis lesions: Implications for the pathogenesis of demyelination. Ann Neurol 2000; 47: 707-71 7

3 Compston A. The genetic epidemiology of multiple sclerosis. Phil Trans R Soc Lond 1999; 354: 1623-1634

4 Coo H, Aronson KJ. A systematic review of several potential nongenetic risk factors for multiple sclerosis. Neuroepidemiology 2004; 23: 1-12

5 McDonald Wl, Compston A, Edan G, et al. Recommended diagnostic criteria for multiple sclerosis: Guidelines from the International Panel on the Diagnosis of Multiple Sclerosis. Ann Neurol 2001; 50: 121-127

6 Steinman L. Multiple sclerosis: A two-stage disease. Nat Immunol 2001; 2: 762-764

7 Trapp BD, Peterson J, Ransohoff RM, et al. Axonal transection in the lesions of multiple sclerosis. N Engl J Med 1998; 338: 278-285

8 Fazekas F, Deisenhammer F, Strasser-Fuchs S, et al. Randomised placebo-controlled trial of monthly intravenous immunoglobulin therapy in relapsing-remitting multiple sclerosis. Austrian Immunoglobulin in Multiple Sclerosis Study Group. Lancet 1997; 349: 589-593

9 Sorensen PS, Wanscher B, Jensen CV, et al. Intravenous immunoglobulin G reduces MRI activity in relapsing multiple sclerosis. Neurology 1998; 50: 1273-1281

10 Sorensen PS, Fazekas F, Lee M. Intravenous immunoglobulin G for the treatment of relapsing-remitting multiple sclerosis: A metaanalysis. Eur J Neurol 2002; 9: 557-563

11 Hurez V, Kazatchkine MD, Vassilev T, et al. Pooled normal human polyspecific IgM contains neutralizing anti-idiotypes to IgG autoantibodies of autoimmune patients and protects from experimental autoimmune disease. Blood 1997; 90: 4004-4013

12 Rossi F, Dietrich G, Kazatchkine M. Anti-idiotypes against autoantibodies in normal immunoglobulins: Evidence for network regulation of human autoimmune responses, lmmunol Rev 1989; 110: 135-149

13 Samuelsson A, Towers TL, Ravetch JV. Anti-inflammatory activity of IVIG mediated through the inhibitory Fc receptor. Science 2001; 291: 484-486

14 Stangel M, Joly E, Scolding NJ, et al. 2000 Normal polyclonal immunoglobulins (IVIg) inhibit microglial phagocytosis in vitro. J Neuroimmunol 2000; 106: 137-144

15 Stangel M, Compston A, Scolding NJ. Oligodendroglia are protected from antibody-mediated complement injury by normal immunoglobulins (IVIg). J Neuroimmunol 2000; 103: 195-201

16 Basta M, Langlois PF, Marques M, et al. High-dose intravenous immunoglobulin modifies complement-mediated in vivo clearance. Blood 1989; 74: 326-333

17 Rodriguez M. Immunoglobulins stimulate central nervous system remyelination: Electron microscopic and morphometric analysis of proliferating cells. Lab Invest 1991; 64: 358-370

18 Warrington AE, Asakura K, Bieber AJ, ef al. Human monoclonal antibodies reactive to oligodendrocytes promote remyelination in a model of multiple sclerosis. Proc Natl Acad Sci USA 2000; 97: 6820-6825

19 Bradl M, Hohlfeld R. Molecular pathogenesis of neuroinflammation. J Neurol Neurosurg Psychiatry 2003; 74: 1364-1370

20 Tanuma N, Shin T, Matsumoto Y. Characterization of acute versus chronic relapsing autoimmune encephalomyelitis in DA rats. J Neuroimmunol 2000; 108: 171-180

21 Lorentzen JC, Issazadeh S, Storch M, et al. Protracted, relapsing and demyelinating experimental autoimmune encephalomyelitis in DA rats immunized with syngeneic spinal cord and incomplete Freund's adjuvant. J Neuroimmunol 1995; 63: 193-205

22 Kornek B, Storch MK, Weissert R, ei ai Multiple sclerosis and chronic autoimmune encephalomyelitis: A comparative quantitative study of axonal injury in active, inactive, and remyelinated lesions. Am J Pathol 2000; 157: 267-276

23 Wujek JR, Bjartmar C, Richer E, et al. Axon loss in the spinal cord determines permanent neurological disability in an animal model of multiple sclerosis. J Neumpathol Exp Neurol 2002; 61: 23-32

24 Pashov A, Dubey C, Kaveri SV, et al. Normal immunoglobulin G protects against experimental allergic encephalomyelitis by inducing transferable T cell unresponsiveness to myelin basic protein. Eur J Immunol 1998; 28: 1823-1831

25 Achiron A, Mor F, Margalit R, et al. Suppression of experimental autoimmune encephalomyelitis by intravenously administered polyclonal immunoglobulins. J Autoimmun 2000; 15: 323-330

26 Kaveri S, Prasad N, Vassilev T, et al. Modulation of autoimmune responses by intravenous immunoglobulin (IVIg). Mult Scler 1997; 3: 121-128

27 Amran D, Renz H, Lack C, et al. Suppression of cytokinedependent human T-cell proliferation by intravenous immunoglobulin. Clin Immunol Immunopathol 1994; 73: 180-186

28 Aktas O, Waiczies S, Grieger U, ef al. Polyspecific immunoglobulins (IVIg) suppress proliferation of human (auto)antigen-specific T cells without inducing apoptosis. J Neuroimmunol 2001; 114: 160-167

29 Andersson UC, Bjork L, Skansen-Saphir U, et al. Down-regulation of cytokine production and interleukin-2 receptor expression by pooled human IgG. Immunology 1993; 79: 211-216

30 Bieber AJ, Warrington A, Pease LR, et al. Humoral autoimmunity as a mediator of CNS repair. Trends Neumsci 2001 ; 24: S39-S44

31 Rodriguez M, Lennon VA. Immunoglobulins promote remyelination in the central nervous system. Ann Neurol 1990; 27: 12-17

32 Stangel M, Boegner F, Klatt CH, et al. Placebo controlled pilot trial to study the remyelinating potential of intravenous immunoglobulins in multiple sclerosis. J Neurol Neurosurg Psychiatry 2000; 68: 89-92

33 Noseworthy JH, O'Brien PC, Petterson TM, et al. A randomized trial of intravenous immunoglobulin in inflammatory demyelinating optic neuritis. Neurology 2001; 56: 1514-1522

34 Sorensen PS, Haas J, Sellebjerg F, et al. IV immunoglobulins as add-on treatment to methylprednisolone for acute relapses in MS. TARIMS study group. Neurology 2004; 63: 2028-2033

35 Achiron A, Gabbay U, Gilad R, et al. Intravenous immunoglobulin treatment in multiple sclerosis. Effect on relapses. Neurology 1998; 50: 398-402

36 Lewanska M, Siger-Zajdel M, Selmaj K. No difference in efficacy of two different doses of intravenous immunoglobulins in MS: clinical and MRI assessment. Eur J Neurol 2002; 9: 565-572

37 Hommes OR, Sorensen PS, Fazekas F, et al. Intravenous immunoglobulin in secondary progressive multiple sclerosis: randomised placebo-controlled trial, tancer 2004; 364: 1149-1156

Signe Humle Jorgensen*, Poul Erik Hyldgaard Jensen*, Henning Laursen[dagger] and Per Soelberg Sorensen*

* Danish MS Centre, Copenhagen University Hospital, Rigshospitalet sect. 9202, DK-2100 Copenhagen, Denmark

[dagger] Laboratory of Neuropathology, Copenhagen University Hospital, Rigshospitalet, DK-2100 Copenhagen, Denmark

Correspondence and reprint requests to: Signe Humle Jorgensen, MS Research Unit, Copenhagen MS Centre, Copenhagen University Hospital, Rigshospitalet sect. 9202, DK-2100 Copenhagen, Denmark. [shumle@nru.dk] Accepted for publication April 2005.

Copyright Maney Publishing Sep 2005
Provided by ProQuest Information and Learning Company. All rights Reserved

Return to Isoflurane
Home Contact Resources Exchange Links ebay