ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Chronic inflammatory demyelinating polyneuropathy: Treatment and prognosis

Chronic inflammatory demyelinating polyneuropathy: Treatment and prognosis
Literature review current through: Jan 2024.
This topic last updated: Jul 06, 2023.

INTRODUCTION — Chronic inflammatory demyelinating polyneuropathy (CIDP; also known as chronic inflammatory demyelinating polyradiculoneuropathy) is an entity that describes a group of related neuropathies, all having chronicity, demyelination, inflammation, and immune mediation in common.

In the classic form of CIDP, motor involvement is greater than sensory. The clinical course of CIDP is slowly progressive in a majority of patients, but a relapsing-remitting course is noted in at least one-third and is more common in the pediatric age group.

CIDP generally responds to immunosuppressive or immunomodulatory treatment with glucocorticoids, intravenous immune globulin, or plasma exchange.

The treatment and prognosis of CIDP will be reviewed here. Other aspects of CIDP are discussed separately. (See "Chronic inflammatory demyelinating polyneuropathy: Etiology, clinical features, and diagnosis".)

GOALS AND COURSE OF TREATMENT — Early administration of effective treatment is important in CIDP. The goal is to stop the immune attack against the myelin sheath of peripheral nerves so that secondary axonal degeneration is minimized [1]. This can improve symptoms and function and can prevent or minimize long-term disability.

The treatment approach depends on the severity and course of disease. While some patients with CIDP have such mild disease with minimal impact on function and quality of life that treatment is not required, most patients are significantly impaired and need treatment. Multiple medication trials may be required to optimize treatment response, particularly for patients with severe disease. Serial clinical examinations are used to monitor for relapses and guide changes in therapy.

Once symptoms stabilize, and for patients who present with milder symptoms, the treatment objective is for sustained improvement and to promote remission.

Consultation with a neuromuscular specialist, if available, is recommended for optimal management.

INITIAL THERAPY — For most treatment-naïve patients with CIDP who are more than mildly affected or for mildly affected patients who are rapidly worsening, we recommend initial immune-modulatory treatment using either intravenous immune globulin (IVIG), plasma exchange, or glucocorticoids [2-4]. These treatments appear to be similarly effective and are considered first-line treatments by guidelines from the European Academy of Neurology and Peripheral Nerve Society [5-8]. The initial choice among these therapies is influenced by disease severity, concurrent illness, venous access, treatment side effects, availability, and cost [9].

Patients with related chronic immune-mediated neuropathies due to nodal or paranodal antibodies (eg, neurofascin, contactin), however, may be unresponsive to typical initial therapies such as IVIG. For these patients, we prefer rituximab or other immunomodulatory therapies. (See "Chronic inflammatory demyelinating polyneuropathy: Etiology, clinical features, and diagnosis", section on 'CIDP variants and related conditions' and 'Patients with refractory disease' below.)

Intravenous immune globulin — IVIG is an effective therapy for CIDP. We start treatment with IVIG when a rapid response is a priority. IVIG is usually easier to administer than plasma exchange and may have a more rapid response for disability improvement than with glucocorticoids.

A number of small randomized controlled trials and systematic reviews have established that IVIG is effective for the short-term treatment of CIDP [3,7,10-15]. In one meta-analysis of five trials with a total of 269 patients, the rate of disability improvement within one month after treatment was significantly higher with IVIG than with placebo (44 versus 26 percent; risk ratio [RR] 2.4, 95% CI 1.72-3.36) [7]. To obtain improvement in one patient, the number needed to treat (NNT) was 3. Overall, IVIG improved disability for at least two to six weeks.

More long-term outcome data are limited. During a 24-week extension phase of the ICE study, 57 initial-phase IVIG responders were randomly reassigned to IVIG or placebo [10]. Patients who continued to receive IVIG had a significantly longer time to relapse than those reassigned to placebo during this phase, and the relapse rate was significantly lower for patients treated with IVIG (13 versus 45 percent, hazard ratio 0.19, 95% CI 0.05-0.70).

The meta-analysis [7] also identified small trials (20 to 46 patients each) that compared IVIG with plasma exchange [5], oral prednisolone [6], and intravenous (IV) methylprednisolone [15]. The benefit of IVIG appeared similar to that of plasma exchange, oral prednisolone, and IV methylprednisolone [7].

Dosing regimens – The initial dose of IVIG is 2 g/kg infused over four to five days (eg, 0.4 g/kg per day for five days). Many patients with CIDP require a treatment trial of repeat IVIG dosing every two to four weeks depending on clinical course.

For most patients, we repeat infusion of IVIG (1 g/kg) every three weeks for two to three months after the initial treatment before determining efficacy. Observation with objective measures should determine if the treatment is effective. Benefit may not be apparent after the first course of IVIG treatment, but 94 percent of IVIG responders in the ICE trial did so within nine weeks [14].

At the completion of the treatment trial, objective signs of efficacy should be evident to continue treatment. However, while progression of disease indicates treatment failure, stability in a patient who previously was progressing may indicate a treatment response and warrant continued treatment for another two to three months before reassessment of treatment efficacy. (See 'Assessing response to therapy' below and 'Inadequate response to initial therapy' below.)

For patients who respond well to therapy, the treatment can be tapered once improvement plateaus. (See 'Maintenance IVIG' below.)

Adverse effectsIVIG use has been associated with headache, nausea, fever, aseptic meningitis, rash, acute renal failure (mostly related to sucrose-containing products), and rarely hyperviscosity and thrombosis. The most common adverse effects from clinical trials on patients with CIDP were transient headache, nausea, and fever [14,16,17]. In a meta-analysis, the risk of serious adverse effects for IVIG was similar to placebo (RR 0.82, 95% CI 0.36-1.87) [7].

Immunoglobulin A (IgA)-deficient patients should not be treated with IVIG since it can lead to anaphylaxis. IVIG should be used cautiously in patients with severe kidney disease because of its association with renal failure [9]. Only formulations without sucrose and with low osmolality should be considered.

Adverse effects of IVIG and strategies to minimize these are reviewed in greater detail separately. (See "Intravenous immune globulin: Adverse effects".)

Plasma exchange — Plasma exchange is an effective alternative to IVIG; however, it may be less convenient to administer. Plasma exchange is available only at specialized centers and has the added concerns of an invasive procedure that requires repeated venous access and (often) indwelling catheters.

Two small randomized clinical trials [18,19], as well as a number of observational studies [20-26], have found that plasma exchange is effective for the short-term treatment of CIDP. In a meta-analysis summarizing the clinical trials, approximately two-thirds of patients treated with plasma exchange showed meaningful short-term clinical improvement as measured by Neuropathy Impairment Score [27].

Like IVIG, a single treatment course with plasma exchange is unlikely to lead to remission. In one of the included trials [19], relapses of CIDP occurred in 8 of 12 patients (67 percent) who initially responded to plasma exchange, most occurring within a week or two after stopping plasma exchange.

Dosing regimens – For patients with CIDP and severe disability, we suggest four to six exchanges over 8 to 10 days. The plasma exchange schedule after that depends on clinical response but can usually be decreased to one exchange every three to four weeks. However, if symptoms occur between treatments, the schedule would be modified to eliminate the interval changes.

The implementation of therapeutic plasma exchange, including techniques and regimens, is discussed in detail separately. (See "Therapeutic apheresis (plasma exchange or cytapheresis): Indications and technology".)

Adverse effects – The main complications of plasma exchange are hypotension, sepsis, and problems with intravenous access. In addition, plasma exchange requires repeated venous access and (often) indwelling catheters that are susceptible to clotting and infection. (See "Therapeutic apheresis (plasma exchange or cytapheresis): Complications".)

Glucocorticoids — For patients with more insidious CIDP, a rapid treatment response may be less critical than for more severely affected patients. For such patients, we suggest initial treatment with glucocorticoids because it is an effective initial treatment strategy that also appears to be effective to help achieve remission [15,28]. In one retrospective study, the median time to relapse was shorter for those treated with IVIG compared with those who received high-dose pulse methylprednisolone (4.5 versus 14 months), although a similar proportion (77 to 86 percent) eventually relapsed in both groups [28].

No large controlled trials of glucocorticoids in CIDP have been performed. Substantial data from retrospective series suggest that oral glucocorticoids are beneficial for CIDP [29-33]. In a small trial, 28 patients completed a three-month regimen of oral prednisone, beginning with a dose of 120 mg every other day during the first week, followed by a slow taper off prednisone by the end of 12 weeks [34]. Patients who received prednisone demonstrated clinically meaningful improvement on disability and electrodiagnostic measures compared with patients assigned to placebo.

Dosing schedule – Glucocorticoids may be administered either by pulse (intermittent) or daily dosing. The selection of treatment regimen involves shared decision-making and should be individualized by patient factors. Both regimens have been shown to be effective and long-term adverse effects appear to be similar, including osteoporosis and diabetes mellitus [35]. However, pulse-dose glucocorticoids may be more likely to cause sleep disturbances and agitation and difficulty with glycemic control for patients with diabetes. Daily dosing may be more likely to cause weight gain and cushingoid features.

For patients treated with pulse-dose glucocorticoids, we typically start with an initial dose of IV methylprednisolone (1000 mg/day) for three days, followed by 1000 mg one day a week for four weeks [36]. Oral pulsed steroids may be an efficacious alternative to an intravenous pulsed regimen. Options include dexamethasone 40 mg for four consecutive days once a month [35] or prednisone 600 mg once a week [37]. In patients who respond to therapy, we taper either the dose or frequency once improvement or clinical stabilization occurs. Earlier tapering and a switch to another agent is necessary if intolerable adverse effects develop.

For patients treated with daily dose glucocorticoids, we typically start prednisone at 1 to 1.5 mg/kg daily (usually around 50 to 80 mg daily; no more than 100 mg). Alternate-day glucocorticoid therapy may reduce the incidence of side effects, but some experts believe it is less effective than a daily dosing regimen [9].

In the PREDICT study, 40 patients with newly diagnosed, treatment-naive CIDP were randomly assigned to treatment with either high-dose pulsed oral dexamethasone (40 mg daily for four days followed by placebo for 24 days, repeated for six cycles) or daily prednisolone (starting with 60 mg daily for five weeks and tapering to alternate-day doses and then to zero over the following 27 weeks) [35]. The remission rate at 12 months was similar between the pulsed high-dose dexamethasone and standard prednisolone treatment groups (10 of 24 [42 percent] versus 6 of 16 [38 percent]; odds ratio 1.19, 95% CI 0.3-4.4).

Route of administration – Whether to use oral or intravenous glucocorticoids is based on patient and physician preference. Oral steroids can be more convenient since intravenous access and nursing support are not necessary. Many patients prefer the convenience of oral therapy. Intravenous therapy may be preferred for patients who may be unable to adhere to a daily medication regimen.

Adverse effects and contraindications – Chronic glucocorticoid use is limited by common and clinically important side effects that include weight gain, cushingoid appearance, easy bruising and skin fragility, cataracts, aseptic necrosis of the femoral or humeral heads, hypertension, diabetes, and osteoporosis. Thus, glucocorticoid therapy is generally contraindicated in patients with peptic ulcer disease, brittle diabetes, refractory hypertension, severe osteoporosis, and systemic fungal infections [9].

Adverse effects of glucocorticoids are discussed in further detail separately. (See "Major adverse effects of systemic glucocorticoids".)

ASSESSING RESPONSE TO THERAPY

Frequency of assessments — Patients with CIDP should be reevaluated routinely (eg, at least every three months when treatments are initiated or changed, and at least twice a year when on maintenance therapy) to determine whether they are responding to therapy. In cases with rapid worsening despite treatment, evaluations at shorter intervals are necessary. For patients in remission, clinical surveillance can occur yearly.

Clinical methods — The success or failure of a treatment regimen should be judged using objective methods both before and during treatment to quantify clinical changes [38,39]. Although such objective measures have not been universally employed in clinical practice, there is accumulating evidence that relying on only the subjective impression of clinical improvement is associated with continuation of ineffective or inappropriate therapy and with the overdiagnosis and excessive treatment of CIDP [39]. By contrast, there may be improvements in energy and well-being with immunomodulatory treatments that do not necessarily point to an effect on the disease itself; it can at times be difficult to stop treatment in these patients because of their misperception of efficacy. (See "Chronic inflammatory demyelinating polyneuropathy: Etiology, clinical features, and diagnosis", section on 'Diagnostic pitfalls'.)

Clinical improvement and disease activity should be assessed and documented using the results of a disability scale and a quantitative grip measure at each appointment. These can be completed in less than five minutes and can be done in the office or by the patient at home.

Commonly used and validated measures of impairment include the Inflammatory Rasch-built Overall Disability Scale (I-RODS) [40,41] and the Inflammatory Neuropathy Cause and Treatment Disability Scale (INCAT) [6]. These measures have been shown to correlate with clinical disease activity [42-44].

Portable devices assessing grip strength dynamometry can provide quantitative grip measures (eg, Jamar or Vigorimeter dynamometer) [10,42,45-47]. The Jamar and Vigorimeter grip devices are relatively inexpensive (<$500 USD).

Additional objective measures to assist in monitoring disease activity and treatment effect include the Timed Up and Go (TUG) test [48], the 10-meter walking test [12], and the Medical Research Council (MRC) sum score [49]. MRC sum score can be obtained by performing a routine neurologic and calculating the summation of MRC grades (0 to 5) given for the following muscle groups (upper arm abductors, elbow flexors, wrist extensors, hip flexors, knee extensors, and foot dorsiflexors); scores range from 0 to 60.

Ultimately, the value of any treatment should include whether the treatment has made a clinically meaningful change (CMC). A CMC threshold has been determined for clinical trials for some of these outcome measures:

I-RODS >4 points in 48-point scale

INCAT >1 point

Grip strength >8 kilopascals or >1.2 pounds/inch2

MRC sum score >3 points

During the course of an initial treatment trial, clinical stability in a patient who was previously progressing or a modest early clinical improvement that does not reach the CMC threshold may nonetheless represent benefit to warrant continued treatment.

The role of electrodiagnostic studies — Under most circumstances, we do not use repeat electrodiagnostic studies to guide treatment decisions.

Testing may be useful in some patients who do not improve clinically in order to assess whether a specific deficit is from prior axon loss or ongoing demyelination. Some clinical deficits due to severe axonal loss may be fixed and unresponsive to immunotherapy. In such patients, the treatment goal may be stabilization of disease rather than rapid improvement, although slow improvement over time related to axonal regeneration is possible. It is important for the clinician and patient to be as clear as possible on what is likely to respond and the anticipated timing of the response.

Some institutions have used summed scores of the compound muscle action potential (CMAP) amplitudes for the motor nerves in the arm and leg to monitor disease activity [50]. We have not found that to be useful in practice.

MANAGING ONGOING THERAPY — The goal for patients with CIDP whose clinical symptoms have stabilized is to promote sustained remission of disease activity.

Some patients will not respond adequately to initial therapy and may benefit from a different therapy or escalation of treatment.

Patients who respond to initial therapy may transition to maintenance therapies.

A small portion of patients (<20 percent) who respond to the initial therapy (treatment until improvement is complete, with slow taper after that) may need no further treatment. However, most patients will relapse either during the taper or after taper is complete.

Inadequate response to initial therapy — If symptoms do not improve or continue to progress after an initial two-to-three-month treatment trial, the patient should be reevaluated to verify the diagnosis of CIDP. (See "Chronic inflammatory demyelinating polyneuropathy: Etiology, clinical features, and diagnosis".)

For patients with CIDP who are nonresponders, a different therapy should be substituted. For those who respond suboptimally to initial therapy, an escalated dose or different therapy should be substituted. As an example, failure to respond to intravenous immune globulin (IVIG) might trigger intervention with plasma exchange and/or immunosuppression with glucocorticoids.

In one series of 67 patients with CIDP, 26 did not respond to initial therapy [26]. Of these, benefit from an alternative treatment was observed in nine (35 percent). For 11 patients who required a third alternative treatment, benefit was observed in three (27 percent).

Managing and preventing relapse — For patients who worsen (during taper or after stopping therapy) after responding to initial therapy, we suggest escalating or repeating the initially successful treatment modality. The timing and dose of ongoing intermittent treatments should be titrated to avoid relapses; this becomes maintenance therapy. Patients treated with IVIG or plasma exchange may require repeated treatment at intervals that typically range from two to six weeks. (See 'Intravenous immune globulin' above and 'Plasma exchange' above.)

If intermittent therapies are insufficient to avoid relapse and/or cannot be tapered, treatment with glucocorticoids or other immunotherapies may be tried to improve the rate of sustained remission. (See 'Maintenance glucocorticoids' below and 'Alternative therapies' below.)

Specific treatment options

Maintenance IVIG — For patients who respond well to initial therapy, we start maintenance intravenous immune globulin (IVIG) at a dose of 1 g/kg, given over one to two days. Subsequent dose titration varies by clinical response. If neuropathy worsens, we increase the maintenance IVIG dose to up to 2 g/kg. For patients who remain clinically stable at 1 g/kg, we taper maintenance IVIG to as low as 0.4 g/kg per dose. In the ProCID trial, 142 patients with CIDP who were treated initially with IVIG at 2 g/kg were assigned to continue maintenance IVIG every three weeks at 0.5 g/kg, 1 g/kg, or 2 g/kg [51]. The proportion of patients with clinical improvement maintained by week 24 was 65 percent in the 0.5 g/kg group, 80 percent in the 1 g/kg group, and 92 percent in the 2 g/kg group. Adverse events were mild and similar between groups. These results suggest that a lowered maintenance dose of IVIG may be effective for many patients, but that a higher dose may be beneficial for others who do not respond to a lower dose. The taper of IVIG dose and/or frequency should be adjusted so that the patient does not deteriorate between doses. We favor tapering the IVIG dose and/or frequency rather than stopping it abruptly. In the PATH study, some of the patients who stopped IVIG did not return back to baseline even after reinstituting treatment [52].

While IVIG therapy can usually control CIDP, most patients require repeated expensive treatments every two to six weeks often for many years; IVIG monotherapy does not usually lead to remission. In a 20-year review of 95 patients treated with IVIG for CIDP, more than 75 percent of the patients improved initially [53]. However, of those who improved, over 85 percent required repeated treatment; fewer than 15 percent were able to discontinue treatment at a mean of 3.5 years and median of 2.1 years. Severity at onset and residual deficit were negative predictors of discontinuation. Maintenance IVIG may be continued in patients who continue to respond and are stable with treatment. When tapering of IVIG leads to worsening of the neuropathy, a long-term immunomodulatory agent can be used as an alternative.

Subcutaneous immune globulin — As an alternative to maintenance IVIG, subcutaneous immune globulin (SCIG) therapy may be used as maintenance therapy for patients with CIDP who respond to IVIG treatment [16,54]. SCIG may be preferred by patients with poor venous access or who live distant from an infusion center. Observational data suggest that SCIG may be better tolerated than IVIG with a reduction in adverse effects including headache and nausea [55,56]. In addition, thrombotic complications seen with IVIG have rarely been seen with SCIG. Although IVIG and SCIG have not been directly compared in clinical trials, they are considered to be similarly effective. SCIG is not approved for use as initial therapy in CIDP.

The PATH trial enrolled 172 adults with CIDP who responded to and were dependent upon IVIG [16]. Subjects were randomly assigned in a 1:1:1 ratio to weekly maintenance treatment with low-dose SCIG (20% solution at 0.2 g/kg), high-dose SCIG (20% solution at 0.4 g/kg), or placebo. At 24 weeks, both doses of SCIG were superior to placebo for reducing the composite risk of a CIDP relapse or withdrawal from the trial for reason other than relapse. Compared with placebo, the absolute risk reduction with low-dose SCIG was 25 percent (95% CI 6-41) and with high-dose SCIG was 30 percent (95% CI 12-46). Adverse events were generally unrelated to SCIG treatment with the exception of one patient who developed an allergic skin reaction.

The administration and dosing of SCIG are reviewed in detail separately. (See "Subcutaneous and intramuscular immune globulin therapy", section on 'Administration and dosing of SCIG'.)

Maintenance glucocorticoids — For most patients who experience ongoing relapses with IVIG or other maintenance therapy, we typically start glucocorticoids. Clinical experience and some studies suggest that glucocorticoid therapy is more likely to produce a clinical remission than either IVIG or plasma exchange. (See 'Glucocorticoids' above.)

Selection among multiple dosing regimens using either oral or intravenous glucocorticoids depends on severity of symptoms, response to treatment and weaning, and patient preference.

Treatment regimens, duration, and tapering – A glucocorticoid course of up to eight weeks may be required to decide whether there is a treatment response [2]. The maximum benefit of glucocorticoid therapy in these studies was seen after one to six months of treatment [9]. However, relapses were common, particularly when tapering the dose [31]. How fast to taper and when to taper also depend on a variety of factors.

Oral – For patients who do not respond to initial therapy, we typically start prednisone at 1 to 1.5 mg/kg daily (usually around 50 to 80 mg daily; no more than 100 mg). For other patients starting glucocorticoids due to a suboptimal response with initial therapy, we typically start prednisone at a lower dose (eg, 20 mg daily). After initiation, the dose is then titrated according to clinical response. Patients who have a good response can begin dose tapering after one to three months.

Glucocorticoid tapering can be done in many ways as a daily or every-other-day regimen. The dose can be tapered by 5 to 10 mg every two to four weeks in clinically stable patients. As an example, with the every-other-day tapering regimen starting from prednisone 80 mg daily:

-Begin with 80 mg alternating with 60 mg for one month, then

-80 mg alternating with 40 mg for one month, then

-80 mg alternating with 0 mg, then

-Taper the every-other-day daily dose by 10 mg each month until 30 mg every other day (or 15 mg daily)

We suggest slowing down the rate of the taper to 5 mg monthly once a dose of 30 mg every other day is reached. It is usually necessary to go back up to a higher dose by at least 10 to 20 mg if symptoms recur during the taper.

Alternatively, pulsed oral steroids at the initial therapeutic dose may be used and tapered every three to four weeks. (See 'Glucocorticoids' above.)

Intravenous – Limited evidence suggests that weekly pulse methylprednisolone (500 mg once a week) is also an effective option for long-term treatment of CIDP [28,37]. Tapering is accomplished by slowly decreasing the frequency of dosing to once every 2 to 12 weeks. As with oral glucocorticoid therapy, there is no standard regimen for pulse intravenous glucocorticoids.

Patients who require sustained doses of prednisone higher than 7 mg daily (or equivalent) to control symptoms may be at elevated risk of chronic adverse effects such as osteoporosis, diabetes, and poor wound healing. For such patients, we typically pursue alternative therapies. (See 'Alternative therapies' below.)

Alternative therapies — Alternative immunomodulatory agents are employed when glucocorticoids are contraindicated or are considered too risky or as steroid-sparing agents. However, the effectiveness of these drugs for CIDP is based on anecdotal reports and small series rather than placebo-controlled trials [57]. They may be useful in patients who do not respond to initial therapy as well as for patients who are refractory to alternative maintenance regimens. (See 'Patients with refractory disease' below.)

The list of immune-modulator drugs that have been used to treat CIDP includes the following:

Azathioprine [32,33,58-60]

Cyclophosphamide [33,61-65]

Cyclosporine [66-69]

Etanercept [70]

Interferon alpha-2a [71,72]

Interferon beta-1a [73,74]

Mycophenolate mofetil [75-77]

Methotrexate [78-80]

Rituximab [81-86]

Tacrolimus [87]

Autologous hematopoietic stem cell transplantation [88,89]

Azathioprine and cyclophosphamide have probably been the most commonly used drugs for CIDP from this list. Azathioprine is generally used as a steroid-sparing medication or reserved for patients with mild disease; a therapeutic response may take over six months. An alternative is mycophenolate mofetil, and clinical experience in patients with myasthenia gravis suggests improvement with this agent may also take a long time.

While many of these agents remain promising, evidence from larger clinical trials is needed to determine whether they are effective for CIDP. Of note, a small randomized placebo-controlled trial in patients with CIDP requiring IVIG or glucocorticoids found no significant benefit for oral methotrexate (15 mg weekly) on any of the primary or secondary outcome measures [79]. However, there was a remarkably high placebo effect, suggesting that the design of the trial may have contributed to the inability to find benefit from the drug. Thus, the role of methotrexate in CIDP remains unproven.

In the rare circumstance that plasma exchange is the only effective therapy, it can be used chronically. However, the treatment brings with it the added risks of sepsis and clotting due to the risk of indwelling catheters.

PATIENTS IN REMISSION — In general, the treatment should be continued until improvement is complete and patients have stabilized (with possible residual deficits). At that point, the treatment can either be tapered or stopped, and clinical surveillance for resurgence of disease activity should continue periodically for several years.

In many cases, appropriate treatment of CIDP with intravenous immune globulin or glucocorticoids leads to improvement or remission within a few months of starting therapy; up to 40 percent of patients will achieve cure (five years or greater stability off treatment) or remission [38,90,91]. Reinstitution of treatment with therapy that had been effective is indicated if the disease becomes active.

PATIENTS WITH REFRACTORY DISEASE — For patients with severe CIDP (eg, unstable active disease with a progressive or relapsing course) who are refractory to treatment with intravenous immune globulin (IVIG), glucocorticoids, and plasma exchange, alternative immunomodulatory treatment options include cyclosporine, methotrexate, rituximab, azathioprine, mycophenolate, and cyclophosphamide (see 'Alternative therapies' above). The choice among these is dependent on a number of factors that include the comfort level of the treating center and clinician with specific agents, side effect profiles of the alternative treatments, disease severity, age and sex of patient, and concurrent medical conditions.

We recommend that patients who are refractory to initial treatment be evaluated at a center with particular expertise in CIDP. The GBS/CIPD Foundation International has a number of designated Centers of Excellence that are available for consultation.

CyclosporineCyclosporine is a treatment for patients with refractory CIDP that appeared effective in one case series of patients with CIDP [68]. Keeping plasma trough levels between 100 and 150 ng/mL may help ensure a therapeutic response [68,69]. Improvement with cyclosporine therapy may occur within three months. However, the side effect profile for cyclosporine is greater than for the other agents. (See "Pharmacology of cyclosporine and tacrolimus", section on 'Side effects'.)

Methotrexate – Another alternative treatment for patients with no contraindications is oral methotrexate (7.5 to 15 mg once a week). Methotrexate was not beneficial for CIDP in a small placebo-controlled randomized trial but the trial had many limitations [79], and methotrexate has appeared to benefit some patients with refractory CIDP [78,80]. It is unclear whether a response to methotrexate occurs within three or four months or takes longer. We suggest at least a four-month trial if the clinical condition allows.

Patients treated with methotrexate should receive folic acid 1 mg daily, or leucovorin 2.5 mg weekly, to prevent hematologic and other side effects. (See "Therapeutic use and toxicity of high-dose methotrexate", section on 'Overview of adverse effects'.)

RituximabRituximab also appears to be effective in patients with refractory CIDP [86]. Additionally, patients with CIDP associated with IgG4 antibodies to neurofascin or contactin may be refractory to IVIG and glucocorticoids but responsive to rituximab [92-94]. Some data also suggest that patients with nodal and paranodal antibodies or hematologic malignancies have a more robust response to treatment [94,95]. Even though this needs further study, rituximab remains a consideration in patients with refractory CIDP. We would consider its use prior to using more aggressive treatments discussed below. (See "Rituximab: Principles of use and adverse effects in rheumatoid arthritis", section on 'Adverse effects'.)

Cyclophosphamide – Pulse intravenous (IV) cyclophosphamide can be effective for patients with severe refractory disease and can lead to long-term remission [33,61,62,64,65]. The usual dosing is 1000 mg/m2 infused monthly for six months. Alternatively, very high-dose (eg, 50 mg/kg administered over one-hour IV for four days) cyclophosphamide regimen may induce a remission of the disease. A systematic review concluded that the available observational studies suggest that cyclophosphamide treatment may be beneficial [57]. These studies included IV pulse regimens and oral daily regimens of cyclophosphamide for patients who failed other therapies. Some of the responses were quite remarkable and sustained [65].

However, cyclophosphamide treatment is associated with potentially life-threatening side effects and should therefore be carefully considered and performed only at specialized centers. (See "General toxicity of cyclophosphamide in rheumatic diseases".)

Autologous hematopoietic stem cell transplantation (AHSCT) – AHSCT has been used as a treatment of last resort for patients with CIDP that is refractory to other therapies, with reported benefit in some cases [88,89]. The main risk associated with AHSCT is infection related to immune suppression; therefore, caution is urged in considering this therapy. Whether AHSCT is more effective than a lower-dose cyclophosphamide regimen is not known. (See "Overview of infections following hematopoietic cell transplantation".)

SPECIAL CIRCUMSTANCES

Pregnancy — The treatment of CIDP in pregnancy is similar to that used for other patients [96]; additional considerations include the following:

Glucocorticoids are frequently used in pregnancy and are considered safe for the fetus.

Plasmapheresis is also considered safe for use in pregnancy, although not without potential complications. (See 'Plasma exchange' above.)

Intravenous immune globulin (IVIG) is generally safe but is associated with potential side effects that might be of particular concern during pregnancy. (See 'Intravenous immune globulin' above.)

The choice among these therapeutic options should be coordinated with the obstetrician. The risks and benefits of these treatments appear to be finely balanced, and the decision is influenced by local availability, clinical expertise, and patient preferences.

Other immunomodulatory drugs discussed above are not recommended for use in pregnancy, particularly early in pregnancy. (See 'Alternative therapies' above.)

Acute infection — Patients with CIDP on chronic immunomodulatory therapy are at an elevated risk of infection. In CIDP patients with severe infection (eg, hospitalized or receiving parenteral antimicrobial therapy), we favor delaying immunomodulatory therapy until infection has come under control. Ideally, such decisions regarding the resumption of therapy are made in consultation with infectious disease specialists.

In general, glucocorticoid therapy should be continued throughout the infection to prevent adrenal insufficiency. (See "Pharmacologic use of glucocorticoids".)

Perioperative issues — No high-quality data or guidelines are available to guide management of CIDP treatment in the perioperative period. Patients with active disease are at risk for clinical worsening in the setting of perioperative systemic stress or exposure to neuromuscular-blocking anesthetic agents. Specific decisions for an individual patient with CIDP undergoing elective surgery are based on disease severity and medical comorbidities and should be planned with the treating surgical team. General principles guiding these decisions include:

For patients with severe or active disease, an escalated dose or timing of current CIDP therapy to pretreat patients prior to elective surgery may be useful.

For patients stable on maintenance therapy, we try to reduce immunosuppressive doses prior to elective surgeries to reduce risks of infection and wound healing issues.

We avoid neuromuscular-blocking agents in these patients. In one retrospective review of 17 patients with CIDP undergoing general anesthesia (including use of succinylcholine in 5 cases and nondepolarizing muscle relaxants in 11 cases), two patients required postoperative mechanical ventilation and three others had clinical worsening [97].

The medical management of glucocorticoids during the perioperative period is discussed separately. (See "The management of the surgical patient taking glucocorticoids".)

PROGNOSIS — Data are limited regarding the long-term prognosis of CIDP. Approximately two-thirds of patients with CIDP will initially respond to any single standard therapy (intravenous immune globulin [IVIG], glucocorticoid, and plasma exchange), while approximately 10 to 15 percent of patients are resistant to all of these [90,98]. Approximately 40 percent of patients with CIDP will achieve cure or remission although may have persistent deficits unresponsive to immunotherapies.

In a systematic review of 21 observational studies that included outcome data on 1199 patients with CIDP, the rate of good functional outcome was 47 percent [91]. These results are limited by heterogeneity in disease severity, treatment, and outcome assessment measures. In a study of 38 Japanese patients with CIDP assessed functional status at five-year follow-up. Patients had been treated with various regimens, including glucocorticoids (89 percent), IVIG (45 percent), and plasma exchange (34 percent). Complete remission was reported in 10 patients (26 percent), partial remission in 23 (61 percent), and ongoing relapses or severe disability in 5 (13 percent). In the partial remission group, there were 13 patients who had stopped immune-modulating treatments, and 10 patients who required intermittent or continuous therapy, mainly glucocorticoids [99].

In a retrospective review of 106 patients with a consensus diagnosis of CIDP and a mean follow-up of 6.4 years who were blindly classified by experts using the CIDP Disease Activity Status (CDAS), the proportion of patients in each category was as follows (table 1) [98]:

Cure (five or more years off treatment): 11 percent

Remission (less than five years off treatment): 20 percent

Stable active disease (one or more years on treatment): 44 percent

Improving (three months to less than one year on treatment): 7 percent

Unstable active disease: 18 percent

SOCIETY GUIDELINE LINKS — Links to society and government-sponsored guidelines from selected countries and regions around the world are provided separately. (See "Society guideline links: Neuropathy".)

SUMMARY AND RECOMMENDATIONS

Goals of treatment – Early, effective treatment is important in chronic inflammatory demyelinating polyneuropathy (CIDP) in order to stop the immune attack against the myelin sheath of peripheral nerves so that secondary axonal degeneration and long-term disability is minimized. (See 'Goals and course of treatment' above.)

Initial immunomodulatory therapy – For treatment-naïve patients with CIDP who are more than mildly affected or for mildly affected patients who are rapidly worsening, we recommend initial immune-modulatory treatment using either intravenous immune globulin (IVIG), plasma exchange, or glucocorticoids (Grade 1B). Each is an effective treatment. (See 'Intravenous immune globulin' above and 'Plasma exchange' above and 'Glucocorticoids' above.)

When rapid response is desired, we suggest initial treatment with IVIG (if available) (Grade 2C). IVIG is usually easier to administer than plasma exchange; both plasma exchange and IVIG may have a more rapid response than that of glucocorticoids.

For patients with more insidious CIDP, we suggest initial treatment with glucocorticoids as an effective initial treatment that is also effective to help achieve remission (Grade 2C). In these patients, a rapid treatment response may be less critical, and glucocorticoids appear to be more effective at achieving remission.

Assessing response to therapy – The success or failure of a treatment regimen should be judged using objective methods using the results of a disability scale and a quantitative grip measure both before and during treatment to quantify clinical changes. (See 'Assessing response to therapy' above.)

Subsequent treatment

Patients with an inadequate response to initial therapy – For patients who fail to respond to the initial mode of therapy, a different therapy should be substituted. For patients who respond suboptimally or worsen (during taper or after stopping therapy) after responding to initial therapy, the initially successful treatment modality is repeated or escalated. (See 'Inadequate response to initial therapy' above and 'Managing and preventing relapse' above.)

Patients who improve or stabilize with initial therapy – For patients who respond well to initial therapy and remain clinically stable, we continue the initial agent as maintenance therapy, tapering as tolerated to prevent relapses. (See 'Specific treatment options' above.)

Patients with relapses – For most patients who experience ongoing relapses with IVIG or other maintenance therapy, we switch to glucocorticoids or an alternative immunotherapy. Glucocorticoid therapy is more likely to produce a clinical remission than either IVIG or plasma exchange. Other immunomodulatory agents may be an alternative but have been less well studied in patients with CIDP. (See 'Maintenance glucocorticoids' above and 'Alternative therapies' above.)

Patients with refractory symptoms – For patients with unstable or progressive symptoms who are refractory to treatment with initial therapy, alternative immunomodulatory treatment options include cyclosporine, methotrexate, azathioprine, mycophenolate, rituximab, and cyclophosphamide. (See 'Patients with refractory disease' above.)

Prognosis – The prognosis of CIDP is generally favorable, but data are limited. Approximately 40 percent of patients achieve cure or remission, while approximately 18 percent have unstable active disease with a progressive or relapsing course. (See 'Prognosis' above.)

  1. Muley SA, Parry GJ. Inflammatory demyelinating neuropathies. Curr Treat Options Neurol 2009; 11:221.
  2. Van den Bergh PY, Hadden RD, Bouche P, et al. European Federation of Neurological Societies/Peripheral Nerve Society guideline on management of chronic inflammatory demyelinating polyradiculoneuropathy: report of a joint task force of the European Federation of Neurological Societies and the Peripheral Nerve Society - first revision. Eur J Neurol 2010; 17:356.
  3. Patwa HS, Chaudhry V, Katzberg H, et al. Evidence-based guideline: intravenous immunoglobulin in the treatment of neuromuscular disorders: report of the Therapeutics and Technology Assessment Subcommittee of the American Academy of Neurology. Neurology 2012; 78:1009.
  4. Oaklander AL, Lunn MP, Hughes RA, et al. Treatments for chronic inflammatory demyelinating polyradiculoneuropathy (CIDP): an overview of systematic reviews. Cochrane Database Syst Rev 2017; 1:CD010369.
  5. Dyck PJ, Litchy WJ, Kratz KM, et al. A plasma exchange versus immune globulin infusion trial in chronic inflammatory demyelinating polyradiculoneuropathy. Ann Neurol 1994; 36:838.
  6. Hughes R, Bensa S, Willison H, et al. Randomized controlled trial of intravenous immunoglobulin versus oral prednisolone in chronic inflammatory demyelinating polyradiculoneuropathy. Ann Neurol 2001; 50:195.
  7. Eftimov F, Winer JB, Vermeulen M, et al. Intravenous immunoglobulin for chronic inflammatory demyelinating polyradiculoneuropathy. Cochrane Database Syst Rev 2013; :CD001797.
  8. Van den Bergh PYK, van Doorn PA, Hadden RDM, et al. European Academy of Neurology/Peripheral Nerve Society guideline on diagnosis and treatment of chronic inflammatory demyelinating polyradiculoneuropathy: Report of a joint Task Force-Second revision. J Peripher Nerv Syst 2021; 26:242.
  9. Ropper AH. Current treatments for CIDP. Neurology 2003; 60:S16.
  10. Hughes RA, Donofrio P, Bril V, et al. Intravenous immune globulin (10% caprylate-chromatography purified) for the treatment of chronic inflammatory demyelinating polyradiculoneuropathy (ICE study): a randomised placebo-controlled trial. Lancet Neurol 2008; 7:136.
  11. Vermeulen M, van Doorn PA, Brand A, et al. Intravenous immunoglobulin treatment in patients with chronic inflammatory demyelinating polyneuropathy: a double blind, placebo controlled study. J Neurol Neurosurg Psychiatry 1993; 56:36.
  12. Thompson N, Choudhary P, Hughes RA, Quinlivan RM. A novel trial design to study the effect of intravenous immunoglobulin in chronic inflammatory demyelinating polyradiculoneuropathy. J Neurol 1996; 243:280.
  13. Hahn AF, Bolton CF, Zochodne D, Feasby TE. Intravenous immunoglobulin treatment in chronic inflammatory demyelinating polyneuropathy. A double-blind, placebo-controlled, cross-over study. Brain 1996; 119 ( Pt 4):1067.
  14. Mendell JR, Barohn RJ, Freimer ML, et al. Randomized controlled trial of IVIg in untreated chronic inflammatory demyelinating polyradiculoneuropathy. Neurology 2001; 56:445.
  15. Nobile-Orazio E, Cocito D, Jann S, et al. Intravenous immunoglobulin versus intravenous methylprednisolone for chronic inflammatory demyelinating polyradiculoneuropathy: a randomised controlled trial. Lancet Neurol 2012; 11:493.
  16. van Schaik IN, Bril V, van Geloven N, et al. Subcutaneous immunoglobulin for maintenance treatment in chronic inflammatory demyelinating polyneuropathy (PATH): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Neurol 2018; 17:35.
  17. Léger JM, De Bleecker JL, Sommer C, et al. Efficacy and safety of Privigen(®) in patients with chronic inflammatory demyelinating polyneuropathy: results of a prospective, single-arm, open-label Phase III study (the PRIMA study). J Peripher Nerv Syst 2013; 18:130.
  18. Dyck PJ, Daube J, O'Brien P, et al. Plasma exchange in chronic inflammatory demyelinating polyradiculoneuropathy. N Engl J Med 1986; 314:461.
  19. Hahn AF, Bolton CF, Pillay N, et al. Plasma-exchange therapy in chronic inflammatory demyelinating polyneuropathy. A double-blind, sham-controlled, cross-over study. Brain 1996; 119 ( Pt 4):1055.
  20. Levy RL, Newkirk R, Ochoa J. Treating chronic relapsing Guillain-Barré syndrome by plasma exchange. Lancet 1979; 2:259.
  21. Server AC, Lefkowith J, Braine H, McKhann GM. Treatment of chronic relapsing inflammatory polyradiculoneuropathy by plasma exchange. Ann Neurol 1979; 6:258.
  22. Gross ML, Thomas PK. The treatment of chronic relapsing and chronic progressive idiopathic inflammatory polyneuropathy by plasma exchange. J Neurol Sci 1981; 52:69.
  23. Toyka KV, Augspach R, Wiethölter H, et al. Plasma exchange in chronic inflammatory polyneuropathy: evidence suggestive of a pathogenic humoral factor. Muscle Nerve 1982; 5:479.
  24. Pollard JD, McLeod JG, Gatenby P, Kronenberg H. Prediction of response to plasma exchange in chronic relapsing polyneuropathy. A clinico-pathological correlation. J Neurol Sci 1983; 58:269.
  25. Pollard JD. A critical review of therapies in acute and chronic inflammatory demyelinating polyneuropathies. Muscle Nerve 1987; 10:214.
  26. Gorson KC, Allam G, Ropper AH. Chronic inflammatory demyelinating polyneuropathy: clinical features and response to treatment in 67 consecutive patients with and without a monoclonal gammopathy. Neurology 1997; 48:321.
  27. Mehndiratta MM, Hughes RA, Pritchard J. Plasma exchange for chronic inflammatory demyelinating polyradiculoneuropathy. Cochrane Database Syst Rev 2015; :CD003906.
  28. Nobile-Orazio E, Cocito D, Jann S, et al. Frequency and time to relapse after discontinuing 6-month therapy with IVIg or pulsed methylprednisolone in CIDP. J Neurol Neurosurg Psychiatry 2015; 86:729.
  29. DeVivo DC, Engel WK. Remarkable recovery of a steroid-responsive recurrent polyneuropathy. J Neurol Neurosurg Psychiatry 1970; 33:62.
  30. Oh SJ. Subacute demyelinating polyneuropathy responding to corticosteroid treatment. Arch Neurol 1978; 35:509.
  31. Wertman E, Argov Z, Abrmasky O. Chronic inflammatory demyelinating polyradiculoneuropathy: features and prognostic factors with corticosteroid therapy. Eur Neurol 1988; 28:199.
  32. Barohn RJ, Kissel JT, Warmolts JR, Mendell JR. Chronic inflammatory demyelinating polyradiculoneuropathy. Clinical characteristics, course, and recommendations for diagnostic criteria. Arch Neurol 1989; 46:878.
  33. McCombe PA, Pollard JD, McLeod JG. Chronic inflammatory demyelinating polyradiculoneuropathy. A clinical and electrophysiological study of 92 cases. Brain 1987; 110 ( Pt 6):1617.
  34. Dyck PJ, O'Brien PC, Oviatt KF, et al. Prednisone improves chronic inflammatory demyelinating polyradiculoneuropathy more than no treatment. Ann Neurol 1982; 11:136.
  35. van Schaik IN, Eftimov F, van Doorn PA, et al. Pulsed high-dose dexamethasone versus standard prednisolone treatment for chronic inflammatory demyelinating polyradiculoneuropathy (PREDICT study): a double-blind, randomised, controlled trial. Lancet Neurol 2010; 9:245.
  36. Lopate G, Pestronk A, Al-Lozi M. Treatment of chronic inflammatory demyelinating polyneuropathy with high-dose intermittent intravenous methylprednisolone. Arch Neurol 2005; 62:249.
  37. Muley SA, Kelkar P, Parry GJ. Treatment of chronic inflammatory demyelinating polyneuropathy with pulsed oral steroids. Arch Neurol 2008; 65:1460.
  38. Cornblath DR, Gorson KC, Hughes RA, Merkies IS. Observations on chronic inflammatory demyelinating polyneuropathy: A plea for a rigorous approach to diagnosis and treatment. J Neurol Sci 2013; 330:2.
  39. Allen JA, Lewis RA. CIDP diagnostic pitfalls and perception of treatment benefit. Neurology 2015; 85:498.
  40. van Nes SI, Vanhoutte EK, van Doorn PA, et al. Rasch-built Overall Disability Scale (R-ODS) for immune-mediated peripheral neuropathies. Neurology 2011; 76:337.
  41. Draak TH, Vanhoutte EK, van Nes SI, et al. Changing outcome in inflammatory neuropathies: Rasch-comparative responsiveness. Neurology 2014; 83:2124.
  42. Draak TH, Pruppers MH, van Nes SI, et al. Grip strength comparison in immune-mediated neuropathies: Vigorimeter vs. Jamar. J Peripher Nerv Syst 2015; 20:269.
  43. Vanhoutte EK, Draak TH, Gorson KC, et al. Impairment measures versus inflammatory RODS in GBS and CIDP: a responsiveness comparison. J Peripher Nerv Syst 2015; 20:289.
  44. Vanhoutte EK, Faber CG, Merkies IS, PeriNomS study group. 196th ENMC international workshop: Outcome measures in inflammatory peripheral neuropathies 8-10 February 2013, Naarden, The Netherlands. Neuromuscul Disord 2013; 23:924.
  45. Vanhoutte EK, Latov N, Deng C, et al. Vigorimeter grip strength in CIDP: a responsive tool that rapidly measures the effect of IVIG--the ICE study. Eur J Neurol 2013; 20:748.
  46. Rajabally YA, Narasimhan M. Jamar hand-held grip dynamometry in chronic inflammatory demyelinating polyneuropathy. J Neurol Sci 2013; 325:36.
  47. Doneddu PE, Hadden RDM. Daily grip strength response to intravenous immunoglobulin in chronic immune neuropathies. Muscle Nerve 2020; 62:103.
  48. Podsiadlo D, Richardson S. The timed "Up & Go": a test of basic functional mobility for frail elderly persons. J Am Geriatr Soc 1991; 39:142.
  49. Kleyweg RP, van der Meché FG, Schmitz PI. Interobserver agreement in the assessment of muscle strength and functional abilities in Guillain-Barré syndrome. Muscle Nerve 1991; 14:1103.
  50. Baek SH, Hong YH, Choi SJ, et al. Electrodiagnostic data-driven clustering identifies a prognostically different subgroup of patients with chronic inflammatory demyelinating polyneuropathy. J Neurol Neurosurg Psychiatry 2019; 90:674.
  51. Cornblath DR, van Doorn PA, Hartung HP, et al. Randomized trial of three IVIg doses for treating chronic inflammatory demyelinating polyneuropathy. Brain 2022; 145:887.
  52. Mielke O, Bril V, Cornblath DR, et al. Restabilization treatment after intravenous immunoglobulin withdrawal in chronic inflammatory demyelinating polyneuropathy: Results from the pre-randomization phase of the Polyneuropathy And Treatment with Hizentra study. J Peripher Nerv Syst 2019; 24:72.
  53. van Doorn PA, Dippel DWJ, van Burken MMG, et al. Longterm IV Immunoglobulin treatment in CIDP. J Periph Nerv Syst 2003; 8 Suppl 1:70.
  54. Goyal NA, Karam C, Sheikh KA, Dimachkie MM. Subcutaneous immunoglobulin treatment for chronic inflammatory demyelinating polyneuropathy. Muscle Nerve 2021; 64:243.
  55. Markvardsen LH, Christiansen I, Andersen H, Jakobsen J. Headache and Nausea after Treatment with High-Dose Subcutaneous versus Intravenous Immunoglobulin. Basic Clin Pharmacol Toxicol 2015; 117:409.
  56. Vu T, Anthony N, Alsina R, et al. Impact of subcutaneous immunoglobulin on quality of life in patients with chronic inflammatory demyelinating polyneuropathy previously treated with intravenous immunoglobulin. Muscle Nerve 2021; 64:351.
  57. Mahdi-Rogers M, Brassington R, Gunn AA, et al. Immunomodulatory treatment other than corticosteroids, immunoglobulin and plasma exchange for chronic inflammatory demyelinating polyradiculoneuropathy. Cochrane Database Syst Rev 2017; 5:CD003280.
  58. Dyck PJ, O'Brien P, Swanson C, et al. Combined azathioprine and prednisone in chronic inflammatory-demyelinating polyneuropathy. Neurology 1985; 35:1173.
  59. Simmons Z, Albers JW, Bromberg MB, Feldman EL. Long-term follow-up of patients with chronic inflammatory demyelinating polyradiculoneuropathy, without and with monoclonal gammopathy. Brain 1995; 118 ( Pt 2):359.
  60. Monaco S, Turri E, Zanusso G, Maistrello B. Treatment of inflammatory and paraproteinemic neuropathies. Curr Drug Targets Immune Endocr Metabol Disord 2004; 4:141.
  61. Prineas JW, McLeod JG. Chronic relapsing polyneuritis. J Neurol Sci 1976; 27:427.
  62. Good JL, Chehrenama M, Mayer RF, Koski CL. Pulse cyclophosphamide therapy in chronic inflammatory demyelinating polyneuropathy. Neurology 1998; 51:1735.
  63. Bouchard C, Lacroix C, Planté V, et al. Clinicopathologic findings and prognosis of chronic inflammatory demyelinating polyneuropathy. Neurology 1999; 52:498.
  64. Brannagan TH 3rd, Pradhan A, Heiman-Patterson T, et al. High-dose cyclophosphamide without stem-cell rescue for refractory CIDP. Neurology 2002; 58:1856.
  65. Gladstone DE, Prestrud AA, Brannagan TH 3rd. High-dose cyclophosphamide results in long-term disease remission with restoration of a normal quality of life in patients with severe refractory chronic inflammatory demyelinating polyneuropathy. J Peripher Nerv Syst 2005; 10:11.
  66. Hodgkinson SJ, Pollard JD, McLeod JG. Cyclosporin A in the treatment of chronic demyelinating polyradiculoneuropathy. J Neurol Neurosurg Psychiatry 1990; 53:327.
  67. Barnett MH, Pollard JD, Davies L, McLeod JG. Cyclosporin A in resistant chronic inflammatory demyelinating polyradiculoneuropathy. Muscle Nerve 1998; 21:454.
  68. Matsuda M, Hoshi K, Gono T, et al. Cyclosporin A in treatment of refractory patients with chronic inflammatory demyelinating polyradiculoneuropathy. J Neurol Sci 2004; 224:29.
  69. Odaka M, Tatsumoto M, Susuki K, et al. Intractable chronic inflammatory demyelinating polyneuropathy treated successfully with ciclosporin. J Neurol Neurosurg Psychiatry 2005; 76:1115.
  70. Chin RL, Sherman WH, Sander HW, et al. Etanercept (Enbrel) therapy for chronic inflammatory demyelinating polyneuropathy. J Neurol Sci 2003; 210:19.
  71. Gorson KC, Ropper AH, Clark BD, et al. Treatment of chronic inflammatory demyelinating polyneuropathy with interferon-alpha 2a. Neurology 1998; 50:84.
  72. Pavesi G, Cattaneo L, Marbini A, et al. Long-term efficacy of interferon-alpha in chronic inflammatory demyelinating polyneuropathy. J Neurol 2002; 249:777.
  73. Hughes RA, Gorson KC, Cros D, et al. Intramuscular interferon beta-1a in chronic inflammatory demyelinating polyradiculoneuropathy. Neurology 2010; 74:651.
  74. Hadden RD, Sharrack B, Bensa S, et al. Randomized trial of interferon beta-1a in chronic inflammatory demyelinating polyradiculoneuropathy. Neurology 1999; 53:57.
  75. Umapathi T, Hughes R. Mycophenolate in treatment-resistant inflammatory neuropathies. Eur J Neurol 2002; 9:683.
  76. Gorson KC, Amato AA, Ropper AH. Efficacy of mycophenolate mofetil in patients with chronic immune demyelinating polyneuropathy. Neurology 2004; 63:715.
  77. Bedi G, Brown A, Tong T, Sharma KR. Chronic inflammatory demyelinating polyneuropathy responsive to mycophenolate mofetil therapy. J Neurol Neurosurg Psychiatry 2010; 81:634.
  78. Fialho D, Chan YC, Allen DC, et al. Treatment of chronic inflammatory demyelinating polyradiculoneuropathy with methotrexate. J Neurol Neurosurg Psychiatry 2006; 77:544.
  79. RMC Trial Group. Randomised controlled trial of methotrexate for chronic inflammatory demyelinating polyradiculoneuropathy (RMC trial): a pilot, multicentre study. Lancet Neurol 2009; 8:158.
  80. Díaz-Manera J, Rojas-García R, Gallardo E, Illa I. Response to methotrexate in a chronic inflammatory demyelinating polyradiculoneuropathy patient. Muscle Nerve 2009; 39:386.
  81. Knecht H, Baumberger M, Tobòn A, Steck A. Sustained remission of CIDP associated with Evans syndrome. Neurology 2004; 63:730.
  82. Briani C, Zara G, Zambello R, et al. Rituximab-responsive CIDP. Eur J Neurol 2004; 11:788.
  83. Gono T, Matsuda M, Shimojima Y, et al. Rituximab therapy in chronic inflammatory demyelinating polyradiculoneuropathy with anti-SGPG IgM antibody. J Clin Neurosci 2006; 13:683.
  84. Gorson KC, Natarajan N, Ropper AH, Weinstein R. Rituximab treatment in patients with IVIg-dependent immune polyneuropathy: a prospective pilot trial. Muscle Nerve 2007; 35:66.
  85. Münch C, Anagnostou P, Meyer R, Haas J. Rituximab in chronic inflammatory demyelinating polyneuropathy associated with diabetes mellitus. J Neurol Sci 2007; 256:100.
  86. Muley SA, Jacobsen B, Parry G, et al. Rituximab in refractory chronic inflammatory demyelinating polyneuropathy. Muscle Nerve 2020; 61:575.
  87. Ahlmén J, Andersen O, Hallgren G, Peilot B. Positive effects of tacrolimus in a case of CIDP. Transplant Proc 1998; 30:4194.
  88. Press R, Askmark H, Svenningsson A, et al. Autologous haematopoietic stem cell transplantation: a viable treatment option for CIDP. J Neurol Neurosurg Psychiatry 2014; 85:618.
  89. Kazmi MA, Mahdi-Rogers M, Sanvito L. Chronic inflammatory demyelinating polyradiculoneuropathy: a role for haematopoietic stem cell transplantation? Autoimmunity 2008; 41:611.
  90. Eftimov F, Vermeulen M, van Doorn PA, et al. Long-term remission of CIDP after pulsed dexamethasone or short-term prednisolone treatment. Neurology 2012; 78:1079.
  91. Al-Zuhairy A, Jakobsen J. Outcome in chronic inflammatory demyelinating polyneuropathy: A systematic review and meta-analysis. Muscle Nerve 2023; 68:388.
  92. Querol L, Nogales-Gadea G, Rojas-Garcia R, et al. Antibodies to contactin-1 in chronic inflammatory demyelinating polyneuropathy. Ann Neurol 2013; 73:370.
  93. Querol L, Nogales-Gadea G, Rojas-Garcia R, et al. Neurofascin IgG4 antibodies in CIDP associate with disabling tremor and poor response to IVIg. Neurology 2014; 82:879.
  94. Querol L, Rojas-García R, Diaz-Manera J, et al. Rituximab in treatment-resistant CIDP with antibodies against paranodal proteins. Neurol Neuroimmunol Neuroinflamm 2015; 2:e149.
  95. Roux T, Debs R, Maisonobe T, et al. Rituximab in chronic inflammatory demyelinating polyradiculoneuropathy with associated diseases. J Peripher Nerv Syst 2018; 23:235.
  96. Kohle F, Kuwabara S, Lehmann HC. Chronic inflammatory demyelinating polyneuropathy and pregnancy: systematic review. J Neurol Neurosurg Psychiatry 2021; 92:473.
  97. Mortenson AR, Sprung J, Watson JC, et al. Chronic inflammatory demyelinating polyradiculoneuropathy and anesthesia: a case series. Acta Neurol Belg 2017; 117:895.
  98. Gorson KC, van Schaik IN, Merkies IS, et al. Chronic inflammatory demyelinating polyneuropathy disease activity status: recommendations for clinical research standards and use in clinical practice. J Peripher Nerv Syst 2010; 15:326.
  99. Kuwabara S, Misawa S, Mori M, et al. Long term prognosis of chronic inflammatory demyelinating polyneuropathy: a five year follow up of 38 cases. J Neurol Neurosurg Psychiatry 2006; 77:66.
Topic 5265 Version 38.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟