You are on page 1of 10

Pain Medicine, 0(0), 2020, 1–10

doi: 10.1093/pm/pnz331
Original Research Article

Downloaded from https://academic.oup.com/painmedicine/advance-article-abstract/doi/10.1093/pm/pnz331/5697247 by University of Waterloo user on 09 January 2020


High-Dose Intravenous Immunoglobulin Is Effective in
Painful Diabetic Polyneuropathy Resistant to Conventional
Treatments. Results of a Double-Blind, Randomized,
Placebo-Controlled, Multicenter Trial
Stefano Jann, MD,* Raffaella Fazio, MD,† Dario Cocito, MD,‡ Antonio Toscano, MD,§
Angelo Schenone, MD,¶ Gerolama Alessandra Marfia, MD,k Giovanni Antonini, MD,kj
Luisa De Toni Franceschini, MD,** Anna Mazzeo, MD,†† Marina Grandis, MD,¶ Daniele Velardo, MD,†
Giorgia Mataluni, MD,k and Erdita Peci, MD‡
*Department of Neurology, Niguarda General Hospital, Milan, Italy; †Department of Neuromuscular Disease, San Raffaele Hospital, Milan, Italy;

Department of Neuroscience, University of Turin, Torino, Italy; §Department of Neuroscience, Psychiatry and Anesthesiology, University of Messina,
Messina, Italy; ¶Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics and Maternal and Child Health, University of Genoa, Genoa,
Italy; kDepartment of Neuroscience, University Tor Vergata, Rome, Italy; kjDepartment of Neuroscience, Mental Health and Sensory Organs, Rome
University “Sapienza,” Sant’Andrea Hospital, Rome, Italy; **Department of Neurology, Alesssandro Manzoni General Hospital, Lecco, Italy;
††
Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy

Correspondence to: Stefano Jann, MD, Department of Neurology, Niguarda Hospital, Piazza Ospedale Maggiore 3, 20162 Mialn, Italy.
Tel: 0264442348; Fax: 0264442819; E-mail: stefano.jann@gmail.com.

Funding sources: This study was funded by Grifols (Barcelona, Spain), manufacturer of Flebogamma DIF 5%.

Disclaimer: Grifols, the funder of the study, had no role in data interpretation or in the decision to submit the manuscript for publication. Grifols sup-
ported reporting of study results by procuring medical writing assistance. All authors had full access to all data in the study and had final responsibility
for the decision to submit for publication.

Conflicts of interest: The authors state that they have no conflicts of interest.

Abstract
Objectives. The efficacy and safety of high-dose intravenous immunoglobulin (IVIG) in treatment-resistant diabetic
painful polyneuropathy (DPN) were assessed. Design. This was a randomized, double-blind, placebo-controlled, mul-
ticenter trial (EudraCT 2010–023883–42). Setting. This trial was conducted at eight sites in Italy with a neurology spe-
cialist level of care. Subjects. Twenty-six diabetic patients with DPN who reported baseline severity of pain >60 units
(mm) on a VAS scale at enrollment and were resistant to antidepressants and antiepileptic drugs were enrolled;
23 were randomized (11 in the IVIG arm and 12 in the placebo arm). All patients completed the study and were evalu-
ated. All patients were Caucasian, 15 were male, and 21 had a diagnosis of type II diabetes. Methods. IVIG (0.4 g/kg/d)
or placebo was given for five consecutive days. Pain intensity (visual analog scale, Neuropathic Pain Symptom
Inventory) and quality of life (36-Item Short-Form Health Survey, Clinical/Patient Global Impression of Change ques-
tionnaires) assessments were performed at visits: baseline, start of therapy (one week later), end of therapy (five
days later), and follow-up (four and eight weeks later). Results. The study achieved its prespecified primary end point
of 50% pain reduction at four weeks after IVIG, achieved in seven of 11 patients (63.6%) in the IVIG group vs zero of
12 in the placebo group (P ¼ 0.0013). Only two adverse events were reported during the study: one patient in the
treatment arm reported a mild “dermatitis psoriasiform,” whereas one patient from the placebo group reported a
mild “influenza.” Conclusions. Treatment with IVIG at the dose given was efficacious and safe for patients with DPN
resistant to standard therapies.

Key Words: Intravenous Immunoglobulin; Diabetic Painful Polyneuropathy; Clinical Trial; Visual Analog Scale (VAS); Neuropathic Pain
Symptom Inventory (NPSI)

C 2020 American Academy of Pain Medicine. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com
V 1
2 IVIG for Painful Diabetic Neuropathy

Introduction we published an open-label trial with IVIG in refractory


Diabetes is the most common cause of neuropathy in pri- neuropathic pain [21]. With this background, it seems
mary care in the Western world, and diabetic neuropathy reasonable to consider a possible beneficial effect of IVIG
in DPN. In this study, the efficacy and tolerability of

Downloaded from https://academic.oup.com/painmedicine/advance-article-abstract/doi/10.1093/pm/pnz331/5697247 by University of Waterloo user on 09 January 2020


(DN) is the most common neurological problem associ-
ated with diabetes [1]. DN is a major cause of disability, IVIG to reduce refractory neuropathic pain in patients
high mortality, and poor quality of life [1]. As the preva- with DPN were assessed.
lence of diabetes increases, so does the prevalence and
morbidity of DN. Although up to 90% of diabetic Methods
patients develop neuropathy [2], only 5.8–34% of DN is
painful [3, 4]. Patients with painful DN (DPN) experi- Study Design and Objectives
ence lower health-related quality of life due to the pain This was a multicenter, randomized (1:1), double-blind,
and its impact on daily functioning in life, and DPN has placebo-controlled phase III clinical trial (EudraCT num-
an increasing burden in terms of health care costs [5]. To ber: 2010–023883–42) to assess the efficacy of high-dose
date, diabetic neuropathic pain represents a therapeutic IVIG (Flebogamma DIF 5%, Instituto Grifols, Barcelona,
challenge because pain relief is often unsatisfactory. Spain) for the treatment of patients with DPN resistant
Despite multiple therapies, 50% of DPN remains severe to standard therapies. The primary objective of the study
[6]. Neuropathic pain is multifactorial and incompletely was to evaluate IVIG efficacy in decreasing the intensity
understood, which is one of the main reasons why its of neuropathic pain. The secondary objectives were to as-
treatment still represents an unmet need. DN results in sess the tolerability of IVIG in the patients taking part in
profound alterations in the function of primary sensory the study and to assess changes in patient quality of life.
neurons and their central projection pathways. Altered This study was conducted in compliance with a clini-
expression of voltage-gated sodium channels in DRG cal protocol approved by the Ethics Committee of
neurons and axons, leading to abnormal currents and in- Niguarda General Hospital in Milan, Italy, regulatory
creased firing of nociceptors, has been found in experi- requirements, good clinical practice (GCP), and the ethi-
mental DPN studies [7, 8]. Recent research suggests that cal principles of the latest revision of the Declaration of
the immune system may also play a significant role, with Helsinki as adopted by the World Medical Association.
reports of overexpression of tumor necrosis factor alpha All participants gave written informed consent before
(TNF-a) and other pro-inflammatory cytokines [9, 10]. participation in the study.
Many of the cytokines and chemokines that coordinate
the cellular response to nerve injury can directly alter the Patient Population
sensory transduction properties of nociceptive afferent Patients were recruited from eight different sites in Italy.
axons, upregulating sodium channels and causing ongo- The inclusion criteria at baseline were 1) male and female
ing activity [11, 12]. In diabetic painful neuropathy, the outpatients aged 18 years; 2) who were willing to pro-
contribution of spinal cord microglia activation to cen- vide written informed consent; 3) with a diagnosis of
tral sensitization is emerging as a new concept. Cytokines DPN of at least six months to a maximum of five years,
released by the microglia can induce central sensitization confirmed as per the Toronto Diabetic Neuropathy
via distinct mechanisms. TNF-a enhances the amplitude Expert Group criteria [22]; 4) with intensity of pain 6
of glutamate-induced excitatory currents, and interleu- points on the visual analog scale (VAS) [23]; 5) with 4
kin-1Beta (IL-1b) increases excitatory synaptic points on the Douleur Neuropathique en 4 Questions
transmission and reduces inhibitory transmission [13]. (DN4) questionnaire [24]; and 6) previously treated with
Streptozotocin (STZ)-induced diabetic rats display an in- at least two lines of treatments (antidepressants [tricyclic
crease in IL-1b and TNF-a expression in the spinal cord antidepressant {TCA} or serotonin-norepinephrine reup-
[14, 15]. take inhibitor {SNRI}], anticonvulsants [gabapentin or
Intravenous immunoglobulins (IVIGs) are an immune- pregabalin], or opioids) that reached therapeutic doses
modulating blood-derived product. The anti- but did not obtain satisfactory results (resistance to treat-
inflammatory effect of IVIGs has been the subject of con- ment: decrease in pain of <30% on the evaluation scale)
siderable investigation. IVIG putative anti-inflammatory or who were unable to take conventional therapies due
mechanisms include blockade of the Fc receptor, en- to intolerable side effects. Paracetamol was the only res-
hancement of antibody catabolism, and the suppression cue therapy permitted. Patients could continue their pre-
of pro-inflammatory cytokines [16]. vious ineffective therapies, and they were aware of the
There are many anecdotal reports about rapid and impossibility of modifying the single doses.
sustained pain relief in patients with chronic inflamma- The exclusion criteria were 1) any condition that, in
tory demyelinating polyneuropathy (CIDP) treated with the investigator’s opinion, might invalidate patient par-
IVIG [17, 18], as well as a few reports supporting the use ticipation in the study; 2) presence of another type of
of IVIGs in complex regional pain syndrome [19] and pain that might create confusion when evaluating painful
other neuropathic chronic pain conditions [20]. Recently, polyneuropathy; 3) having been treated with IVIG in the
Jann et al. 3

six months before the start of the study; 4) IgA serum screening visit, the investigators recorded all the informa-
concentration <70 mg/dL; 5) decompensated cardiovas- tion regarding patients’ medical/neurological history, as
cular disease that was not pharmacologically controlled, well as details about personal characteristics (age, sex,
renal insufficiency, hepatic insufficiency, severe altera- weight, height), duration of DN glucose control, previous

Downloaded from https://academic.oup.com/painmedicine/advance-article-abstract/doi/10.1093/pm/pnz331/5697247 by University of Waterloo user on 09 January 2020


tions in blood cell count (leukopenia, thrombocytopenia, analgesic therapies, and ongoing therapies for concomi-
pancytopenia) or psychiatric disturbances; 6) amputa- tant pathologies.
tions; 7) pregnancy and lactation; and 8) participation in At Visit 2, patients eligible to start the treatment were
other clinical studies during the 30-day period before the treated with placebo (intravenous [IV] saline) or IVIG.
start of this study. Regardless of randomization, patients continued taking
In a second phase of inclusion, patients meeting all the other medications at unchanged doses for the remainder
above-mentioned inclusion and none of the exclusion crite- of the study.
ria at baseline were asked to complete the VAS test daily Patients enrolled in the IVIG treatment group received
and record the results in a diary. To be eligible to start the Flebogamma 5% DIF at 0.4 g/kg/d for five consecutive
treatment (Visit 2), patients had to have a mean of 6 cm days (total dose 2 g/kg) in addition to their regular drug
on the VAS test in the seven days after the baseline visit therapy. The study product was infused IV at an initial
and have completed at least 50% of their diary entries. rate of 0.01–0.02 mL/kg/min during the first 30 minutes.
If well tolerated (absence of adverse reaction associated
with the infusion), the administration rate was gradually
Randomization and Masking
increased to a maximum of 0.1 mL/kg/min.
Assignment of patients to the treatment groups (IVIG/
Data from the study were recorded by the investigator
placebo) was carried out by means of a randomization
in electronic case report forms (e-CRFs) while ensuring
list generated by an independent, unmasked statistical
patient anonymization.
team at a contract research organization (Sintesi
Research S.r.l.). Block randomization was used to bal-
ance the quantitative asymmetry of patients assigned to Sampling and Pain Testing
the groups both during and after enrolment. The investi- Blood samples were drawn at baseline to assess IgA se-
gators were not informed about the size of the blocks. rum concentration (mg/dL).
The study was in a double-blind format: Throughout the The following pain intensity and quality of life assess-
study, the patients, physicians, and staff members in- ments were performed at every visit: a) VAS [23], b)
volved (co-investigators, nurses, laboratory staff) and Italian version of Neuropathic Pain Symptom Inventory
assessors did not know which treatment was allocated, (NPSI) [25], and c) 36-Item Short-Form Health Survey
so that they could not be influenced by that knowledge. (SF-36) [26, 27].
The randomization codes were not open until all the data Patients were also asked to complete the VAS scale daily
were entered and the database was sealed, except in the and record the results in a diary during the window be-
case of a severe adverse event presumably related to the tween Visits 1–2 (seven days, week 1), Visits 2–3 (five days,
treatment product. week 2), Visits 3–4 (one month, weeks 3, 4, 5, and 6), and
Blue opaque bags and yellow infusion kits were used Visits 4–5 (one month, weeks 7, 8, 9, and 10). Throughout
for the blind procedure. The blue bags had corresponding the study, four diaries covering a total of 10 weeks were
labels that specified the patient for whom they were handed out to each patient at Visits 1, 2, 3, and 4.
intended. The blinding procedure was carried out at the Additionally, at Visits 3, 4, and 5, both the patient
same centers by qualified nurses who checked the drug and the physician had to complete the Clinical/Patient
vials being dispensed from the pharmacy service and fit- Global Impression of Change (CGIC/PGIC) question-
ted the vials into the blue bags. The nurses were commit- naires [28].
ted to concealing the identity of treatments from
investigators and patients involved in the study. These
Efficacy Assessments
nurses were not otherwise involved in the trial.
The primary efficacy end point was the proportion of
patients with pain reduction 50% of the baseline value.
Study Procedures and Infusions These changes in pain relief were assessed by VAS scale
The duration of the study was 12 weeks and included five as the average of the last seven days registered in the pa-
visits: Visit 1 (baseline), Visit 2 (start of therapy, one tient diary before Visit 4 (four weeks after end of ther-
week after Visit 1), Visit 3 (end of therapy, five days after apy) and before treatment (Visit 2).
Visit 2), Visit 4 (follow-up, four weeks after Visit 3), and Secondary efficacy end points were evaluating the in-
Visit 5 (follow-up, eight weeks after Visit 3). tensity of pain (assessed by the VAS scale, taken from
During the baseline visit (Visit 1), eligible patients both the patient diary and assessed at each visit); identify-
meeting all the inclusion and none of the exclusion crite- ing the type of pain using the NPSI questionnaire (overall
ria were randomized (1:1) to either the IVIG-treated score and single question scores), evaluated at the three
group or the placebo/control group. During this control visits; assessing quality of life using the SF-36
4 IVIG for Painful Diabetic Neuropathy

Health Survey questionnaire (by considering the SF-36 one infusion of IVIG/placebo, whereas the population
modules: physical function, role and physical health, used for the efficacy analysis included all enrolled
pain, general health, energy/vitality, social functioning, patients with at least one infusion of IVIG/placebo and at
role-mental, mental health, and changes in health status); least one follow-up efficacy analysis.

Downloaded from https://academic.oup.com/painmedicine/advance-article-abstract/doi/10.1093/pm/pnz331/5697247 by University of Waterloo user on 09 January 2020


and evaluating overall clinical judgment, assessed by the For qualitative variables, the absolute number and
physician and the patient (CGIC/PGIC questionnaire). proportions are provided. For quantitative variables, the
The mean VAS results obtained at Visits 3, 4, and 5 mean, range, SD of the mean, and 95% confidence inter-
for the IVIG-treated and placebo groups were compared val (CI) of the mean are presented.
against their corresponding mean VAS scores obtained at For a pain intensity reduction 50% (the primary effi-
Visit 2. The weekly means of the VAS daily scores were cacy parameter), the two treatment groups were com-
used to evaluate the pain decrease in each treatment pared by chi-square test (Fisher exact test). Moreover,
group, comparing the values obtained during the week the mean values of the percent pain reduction of the two
before Visit 4 and Visit 5 with the mean value registered treatment groups were compared using the Wilcoxon
the last seven days before Visit 2. rank-sum test. Secondary efficacy parameters (intensity
The mean NPSI results obtained for each individual of pain as assessed by the VAS scale, SF-36 modules,
question at Visits 3, 4, and 5 for the IVIG-treated and NPSI questionnaire) were analyzed as changes vs baseline
placebo groups were compared against their correspond- values by Wilcoxon rank-sum test at each visit. The over-
ing mean NPSI individual question scores obtained at all clinical judgment, assessed by the physician and the
Visit 2. Also, the Total NPSI score for each visit was cal- patient, of the two treatment groups was compared by
culated as the sum of the corresponding 12 questions’ chi-square test.
(Q1 to Q12) mean scores. Statistical significance was set at P  0.05. Analysis
For the SF-36 questionnaire, the influence of the treat- was performed using SAS software, version 9.2 (SAS
ment in the nine modules evaluated was assessed: The Institute Inc., Cary, NC, USA). The contract research or-
mean value per module recorded during Visits 3, 4, and 5 ganization (Sintesi Research S.r.l.) oversaw the study and
was compared against the specific module results analyses.
obtained during Visit 2. Additionally, randomized
patients who received at least one dose of the study drug
were asked to self-assess for each visit their general
Results
health status (HS) as “a little better than one year ago”/ Patients and IVIG Administration
“approximately equal to one year ago”/“a little worse A total of 26 patients were screened at the baseline visit.
than one year ago”/“much worse than one year ago.” Three patients were eligible but failed before starting treat-
The CGIC/PGIC questionnaire was performed during ment (Visit 2): One declined to participate, and two did not
Visits 3, 4, and 5, and the proportions obtained for each meet the additional inclusion criterion (reaching a mean of
study arm were evaluated for statistical significance. 6 cm on the VAS test in the seven days after the baseline
visit). The remaining 23 patients were randomized to the
treatment group (N ¼ 11) or the placebo group (N ¼ 12).
Safety Assessments
All patients completed the study. All patients completed the
All adverse events (AEs) occurring for a participating pa-
study and were evaluated for efficacy and safety. The flow
tient from the first IVIG infusion to the end of the study pe-
of patients is shown in Figure 1.
riod were registered, along with the number and percentage
Most patients were male (N ¼ 15, 65.2%), Caucasian
of patients with each type of event. The severity of AEs was
(100%), and had a diagnosis of type II diabetes (N ¼ 21),
classified as “mild,” “moderate,” or “severe” in accordance
with age between 28 and 77 years, weight at baseline be-
with the protocol. Based on the investigator’s opinion, the
tween 63 and 112 kg, IgA level between 70 and 497 mg/
relationship of the AE with the study drug was classified as
mL, and severity of pain at baseline between 7 and
“not associated,” “improbable,” “possible,” “probable,”
10 cm. No statistically significant differences between the
or “definitely related.”
treatment and placebo groups were found for baseline
characteristics (Table 1).
Statistical Analysis Previous analgesic therapies (before screening visit) in
The study was initially planned to enroll 54 patients in both patient groups included antidepressants (N ¼ 15,
total, to have a 90% power to detect a difference be- 65.2%), opioids (N ¼ 10, 43.5%), and anticonvulsants
tween the IVIG-treated group and the placebo group at a (N ¼ 17, 73.9%). One patient (4.4%) in the IVIG group
significance alpha level of 0.01. However, to compensate took nonsteroidal anti-inflammatory drugs (NSAIDs).
for patients who might drop out, this sample size was in- During the study, seven patients (63.6%) from the
creased by 30%; therefore, 70 patients (35 for each study IVIG group and seven patients (58.3%) from the placebo
group) were expected to enroll in the study. group were treated with at least with one concomitant
The safety population used for the safety analysis con- analgesic therapy (including ongoing medication at base-
sisted of all enrolled subjects who had received at least line visit): opioids (N ¼ 6, 26.1%, four from the IVIG
Jann et al. 5

Enrolled patients
(n= 26)
Not included (n= 3)
• Screening failure (n= 2)
• Withdrawn consent (n= 1)

Downloaded from https://academic.oup.com/painmedicine/advance-article-abstract/doi/10.1093/pm/pnz331/5697247 by University of Waterloo user on 09 January 2020


Randomized
(n= 23)

Treatment Placebo
(n= 11) (n= 12)

Discontinued Discontinued
(n= 0) (n= 0)

Completed and Completed and


evaluable evaluable
(n= 11) (n= 12)
Figure 2. Pain–visual analog scale (VAS; cm). Visit assessment.
Figure 1. Flow of patients through the study. Changes vs start of the therapy (Visit 2). N ¼ 11–12 (intravenous
immunoglobulin–placebo); Mean 6 95% confidence interval;
*P < 0.05.
Table 1. Demographics and clinical characteristics at baseline

Characteristics IVIG (N ¼ 11) Placebo (N ¼ 12) group and zero in the placebo group (P ¼ 0.0013). The
Sex, male, No. (%) 7 (63.6) 8 (66.7) mean percent pain decrease at four weeks was significant,
Race, Caucasian, No. (%) 11 (100) 12 (100) in favor of IVIG treatment vs placebo (–44.2% 6 34.5%
Age, median (range), y 63 (32–70) 64 (28–77) vs –5.2% 6 12.0%, P  0.027).
Baseline weight, mean 6 SD, kg 88.4 6 15.7 81.3 6 14.3
Diabetes type I, No. (%) 1 (9.1) 1 (8.3)
Diabetes type II, No. (%) 10 (90.9) 11 (91.7) Secondary Efficacy Analysis
Duration of diabetes, median (range), y 11.5 (1–23) 15 (4–30)
Duration of DPN, median (range), y 2.6 (1–5) 2.4 (0.5–4)
The VAS score values at Visit 2 were 8.7 6 1.1 and
VAS score at baseline, mean 6 SD 8.4 6 1.1 8.9 6 1.1 9.3 6 1.0 for the IVIG and placebo groups, respectively.
IgA, mean 6 SD, mg/dL 181.8 6 114.2 176.5 6 128.0 The score changes with respect to Visit 2 observed in
Medication failures before the study Visits 3–5 are shown in Figure 2. The difference was sta-
Opioids, No. (%); MME 5 (45.5); 60 5 (41.7); 60 tistically significant at Visits 3 (P ¼ 0.032) and 4
Anticonvulsants, No. (%) 7 (63.6) 10 (83.3)
Antidepressants, No. (%) 7 (63.3) 8 (66.7)
(P ¼ 0.048). A similar trend was observed for the VAS
Other drugs, No. (%) 1 (9.1) 0 score change obtained from pain intensity registered in
Cointerventions during the study the patient diary at Visit 4 (–4.20 6 3.32 vs –0.46 6 1.01
Opioids, No. (%); MME 4 (36.4); 60 2 (16.7); 60 for the IVIG and placebo groups, respectively, P ¼ 0.035)
Anticonvulsants, No. (%) 0 6 (51.0) and Visit 5 (–3.66 6 3.43 vs –0.22 6 0.94 for the IVIG
Antidepressants, No. (%) 3 (27.3) 3 (25.5)
Other drugs, No. (%) 6 (54.5) 4 (33.3)
and placebo groups, respectively, P ¼ 0.078).
The NPSI total score values at Visit 2 were 8.6 6 1.4
DPN ¼ diabetic peripheral neuropathy; MME ¼ morphine milligram and 9.0 6 0.6 for the IVIG and placebo groups, respec-
equivalents, average daily dose; VAS ¼ visual analog scale (intensity of pain). tively. The score change with respect to Visit 2 observed
in Visits 3–5 is shown in Table 2. The difference was sta-
group and two from the placebo group), antidepressants tistically significant for all three visits (P ¼ 0.014,
(N ¼ 6, 26.1%, three from each group), and anticonvul- P ¼ 0.002, and P ¼ 0.018, respectively). Regarding the
sants (N ¼ 9, 39.1%, six from the IVIG group and three NPSI single questions, the score change with respect to
from the placebo group). The most widely used nonanal- Visit 2 was statistically significant, in favor of IVIG treat-
gesic concomitant medications were blood glucose–low- ment (Table 2).
ering drugs, excluding insulin, administered in 14 out of The comparison of SF-36 scores evidenced improve-
23 patients (seven patients in both treatment groups), ment in favor of IVIG treatment. For the general health
and insulin and analogues, administered in nine out of 23 module, the score change difference was statistically sig-
patients (four patients in the IVIG group and five in the nificant with respect to Visit 2 in all three subsequent vis-
placebo group). its (P ¼ 0.008 at Visit 3, P ¼ 0.015 at Visit 4, and
P ¼ 0.014 at Visit 5) (Table 3). Regarding the other mod-
Primary Efficacy Analysis ules, pain, mental health, role and physical health,
The number of patients at Visit 4 (four weeks after end mental health, energy/vitality, social functioning, role-
of therapy) with a pain intensity decrease 50% with re- mental, and physical function, at Visit 3 P was nonsignif-
spect to the baseline level was seven (63.6%) in the IVIG icant. At Visits 4 and 5, the score change was statistically
6 IVIG for Painful Diabetic Neuropathy

0.08 6 0.90
0.17 6 1.03
0.17 6 0.94
–0.33 6 1.07
–0.08 6 1.16
–0.08 6 0.79
–0.17 6 1.40
–0.50 6 1.93
–0.08 6 0.90
0.50 6 1.62
–0.33 6 0.78
–0.08 6 0.90
0.33 6 7.78
Table 2. Neurophatic Pain Symptom Inventory (NPSI). Score change versus Visit 2 (start of the therapy) of Visit 3 (end of therapy), Visit 4 (4-week follow-up) and Visit 5 (8-week follow- significant for all modules (Table 3) except physical func-
tion (nonsignificant at Visit 4) and role and physical

Placebo
health (statistically significant at both Visits 4 and 5).
The SF-36 Change in Health Status survey also

Downloaded from https://academic.oup.com/painmedicine/advance-article-abstract/doi/10.1093/pm/pnz331/5697247 by University of Waterloo user on 09 January 2020


revealed a statistically significant differences in favor of
IVIG treatment at Visits 3 (P ¼ 0.022), 4 (P ¼ 0.022), and
Visit 5 vs Visit 2

–3.91 6 3.08**

–3.91 6 3.94**

–4.73 6 1.95**
–3.82 6 2.75**
–3.27 6 3.29**
–4.27 6 3.52**
–3.73 6 2.76**
–3.64 6 4.32*

–39.7 6 33.5*
–2.64 6 4.59 5 (P ¼ 0.028) (Figure 3).
–2.64 6 4.50

–1.55 6 1.69
–1.64 6 1.96
Concerning the CGIC/PGIC questionnaires, a statisti-
cally significant difference in favor of IVIG treatment
IVIG

was obtained at all visits in both the physician (Visit 3,


P ¼ 0.013; Visit 4, P ¼ 0.008; Visit 5, P ¼ 0.003) and pa-
tient assessments (Visit 3, P ¼ 0.006; Visit 4, P ¼ 0.010;
–0.75 6 0.97
–0.33 6 0.65
–0.25 6 1.14
–0.67 6 0.65
–0.25 6 1.14
–0.08 6 1.08
–0.75 6 1.48
0.50 6 1.93
–0.67 6 1.44
0.08 6 1.83
0.17 6 0.72
–0.25 6 0.87
–3.58 6 6.26
Visit 5, P ¼ 0.003) (Figure 4).
Placebo

Safety Analysis
Only two AEs were reported during the study: One pa-
tient in the treatment arm reported a mild “dermatitis
Visit 4 vs Visit 2

psoriasiform,” judged by the investigator as possibly re-


–6.00 6 1.95**
–4.91 6 2.59**
–4.09 6 2.66**

–3.72 6 2.41**

–47.9 6 27.0**
–3.64 6 3.23*
–4.00 6 3.35*
–4.18 6 3.34*
–4.18 6 3.52*

–4.82 6 3.09*

–1.73 6 1.49*
–1.91 6 1.58*
–3.91 6 3.05

lated to the study treatment, whereas one patient from


the placebo group reported a mild “influenza,” judged by
IVIG

the investigator as not related to the study treatment


(Table 4).
–1.00 6 0.95
–1.00 6 1.28
–0.83 6 1.19
–1.58 6 1.31
–0.67 6 1.97
–1.25 6 0.75
–0.92 6 1.62
–0.42 6 1.56
–1.25 6 1.54
–0.08 6 1.83
–0.33 6 0.65
–0.25 6 0.62
–9.92 6 11.0

Discussion
Placebo

This is the first randomized, double-blind, placebo-con-


trolled clinical trial exploring the possible beneficial ef-
fect of IVIG in DPN. Even with a relatively small sample
size, our results showed statistically significant improve-
Visit 3 vs Visit 2

–3.73 6 2.69*

–3.27 6 2.45*
–3.73 6 2.49*
–3.73 6 2.76*
–3.64 6 1.96*
–3.00 6 2.32*
–2.73 6 1.74*
–3.18 6 2.79*
–0.42 6 1.62*
–1.45 6 1.29*
–1.36 6 1.29*
–35.4 6 19.2*

ment in favor of IVIG in both primary and secondary


–2.64 6 2.38

outcomes.
All patients were diabetics with a long history of pain-
IVIG

ful neuropathy. According to the Toronto Diabetic


Neuropathy Expert Group, the diagnosis of DPN is a
IVIG ¼ intravenous immunoglobulin; NPSI ¼ Neuropathic Pain Symptom Inventory.

clinical one, which relies on the patient’s description of


9.00 6 0.60
8.42 6 1.62
8.33 6 2.10
8.83 6 0.83
8.17 6 1.64
8.83 6 1.70
9.17 6 1.11
8.17 6 2.08
8.75 6 1.06
8.83 6 1.70
4.83 6 0.39
4.67 6 0.78
96.0 6 12.0

pain, hyperalgesia, and frequently allodynia upon exami-


Placebo

nation [22]. Importantly, all centers involved in this


study had neurologists trained in painful neuropathies.
The primary efficacy end point evidenced that IVIG
was highly effective in decreasing pain intensity in DPN
patients. After treatment, almost two-thirds of patients
8.64 6 1.36
6.73 6 3.47
7.00 6 2.90
8.27 6 1.79
7.91 6 2.21
8.45 6 2.58
8.27 6 2.05
7.18 6 3.34
7.91 6 2.30
8.55 6 1.51
4.73 6 0.47
4.27 6 1.12
87.9 6 20.6

showed that pain intensity decreased by half or more


Visit 2

compared with baseline intensity. All secondary end


IVIG
up). N ¼ 11–12 (IVIG-Placebo); Mean 6 SD

points supported this finding. Information from the NPSI


questionnaire, particularly, allowed discriminating and
quantifying five distinct sorts of pain (Q1, Q2, Q3, Q5,
Q5: Is the pain like an electric shock?

and Q6) and feelings (Q11 and Q12), as well as measur-


Q11: Do you feel pins and needles?
Q2: Is it a sort of tightening pain?

Q4: Spontaneous pain (past 24 h)


Q3: Is it a sort of squeezing pain?

Q8: Pain increased by brushing?

ing changes due to IVIG treatment (Q8 to Q10, in all


Q9: Pain increased by pressure?
Q10: Pain increased by cold?

three questions with very significant improvement).


Q7: Pain attacks (past 24 h)
Q12: Do you feel tingling?
Q6: Is it a stabbing pain?

Response to IVIG was consistent, in contrast to the de-


Q1: Is it a burning pain?

*P < 0.05; **P < 0.01.

scribed heterogeneity in response to duloxetine/pregaba-


lin treatment in patients with DPN, associated with
distinct pain characteristics at baseline [29].
Total score
NPSI Item

The rate of improvement that we observed is one of


the highest in neuropathic pain trials. Moreover, it is un-
usual to reach such significance in this setting (double-
Jann et al. 7

Table 3. Change versus Visit 2 (start of the therapy). Score change versus Visit 2 (start of the therapy) of Visit 3 (end of therapy),
Visit 4 (4-week follow-up) and Visit 5 (8-week follow-up). N ¼ 11–12 (IVIG-Placebo); Mean 6 SD

Visit 2 Visit 3 vs Visit 2 Visit 4 vs Visit 2 Visit 5 vs Visit 2

Downloaded from https://academic.oup.com/painmedicine/advance-article-abstract/doi/10.1093/pm/pnz331/5697247 by University of Waterloo user on 09 January 2020


SF-36 Item IVIG Placebo IVIG Placebo IVIG Placebo IVIG Placebo
Physical function 30.5 6 24.3 23.8 6 25.8 4.55 6 11.06 –0.83 6 19.4 4.6 6 11.1 –0.8 6 19.4 30.9 6 27.4* –2.08 6 30.5
Role and physical health 9.09 6 23.1 0.00 6 0.00 15.9 6 32.2 0.00 6 0.00 15.9 6 32.2 0.00 6 0.00 54.6 6 49.8 10.5 6 29.1
Pain 7.7 6 13.2 5.50 6 9.95 18.3 6 16.4 5.25 6 8.30 18.3 6 16.4** 5.25 6 8.30 45.6 6 27.7** 2.42 6 5.96
General health 14.1 6 17.3 9.875 6 15.2 16.2 6 13.6** 0.67 6 7.50 16.2 6 13.6* 0.67 6 7.50 40.8 6 34.8* –0.83 6 8.21
Energy/vitality 33.2 6 10.6 35.0 6 12.6 9.09 6 9.17 6.25 6 13.0 9.09 6 9.17** 6.25 6 13.0 20.0 6 18.2* 2.08 6 8.38
Social functioning 17.1 6 33.7 11.5 6 18.8 12.5 6 18.5 6.25 6 11.3 12.5 6 18.5* 6.25 6 11.3 43.2 6 43.4* 4.17 6 17.1
Role-mental 9.09 6 15.6 16.7 6 33.3 9.09 6 39.7 0.00 6 14.2 9.09 6 39.7* 0.00 6 14.2 66.7 6 51.6* 5.56 6 13.0
Mental health 32.0 6 10.3 34.0 6 11.0 10.6 6 13.5 0.17 6 8.29 10.6 6 13.5** 0.17 6 8.29 32.0 6 10.3* –2.67 6 9.08

IVIG ¼ intravenous immunoglobulin; SF-36 ¼ 36-Item Short-Form Health Survey.


*P < 0.05; **P < 0.01.

100
* * *

80 Much better
Percentage

A little better
60
Approximately equal

40 A little worse
Much worse
20

0
Treatm ent: IVIG Plac IVIG Plac IVIG Plac IVIG Plac IVIG Plac
Visit: 1 (baseline) 2 3 4 5

Figure 3. Thirty-Six-Item Short-Form Health Survey. Change in health status (intravenous immunoglobulin [IVIG]–placebo). For
Visits 1 and 2, patients were asked to self-assess their general health status as “a little better than one year ago”/“approximately
equal to one year ago”/“a little worse than one year ago”/“much worse than one year ago” (see the Methods section). Percentage
of patients; N ¼ 11–12 (IVIG-placebo); *P < 0.05.

100
* ** ** ** * **

80 Much improved
Percentage

Moderately improved
60
Slightly improved

40 No changes
Slightly worsened
20

0
IVIG Plac IVIG Plac IVIG Plac IVIG Plac IVIG Plac IVIG Plac
Tratment:
Visit: 3 4 5 3 4 5

Assessment: By physician By patient

Figure 4. Overall clinical judgment. Physician and patient assessment (intravenous immunoglobulin [IVIG]–placebo). Percentage of
patients; N ¼ 11–12 (IVIG-placebo); *P < 0.05; **P < 0.01.

blind clinical trials in painful neuropathy). In reporting a “the best treatment is only slightly better than the
recent systematic review and meta-analysis of pharmaco- worst.”
logic therapies for DPN that included 58 studies and We are aware that a significant P value does not al-
11,883 patients, Snedecor et al. [30] observed that de- ways have clinical relevance or mean that the trial is posi-
spite the large number of treatments available, the rela- tive. A limitation of this trial was that the recruitment
tive equivalence of their treatment effects means that target was not met due to extremely selective inclusion
8 IVIG for Painful Diabetic Neuropathy

Table 4. Adverse events

Treatment Group Adverse Event Severity Relation with Study Product


IVIG Dermatitis psoriasiform (N ¼ 1) Mild Possible

Downloaded from https://academic.oup.com/painmedicine/advance-article-abstract/doi/10.1093/pm/pnz331/5697247 by University of Waterloo user on 09 January 2020


Placebo Influenza (N ¼ 1) Mild No relation

criteria, and this can be considered a weakness of the some chronic pain conditions. IVIG can significantly re-
study. The main problem for enrollment was the absence duce pro-inflammatory cytokine levels. Sharief et al. [40]
of renal involvement. Nevertheless, enrolled patients showed that in patients with Guillain-Barre syndrome,
were all resistant to conventional treatments or com- TNF-a and IL-1b decreased on day 3 and significantly by
plained of intolerable side effects. They had poor quality day 5 after IVIG infusion. IL-10 and IL-2 were not af-
of life and were exhausted by intractable pain. fected by IVIG treatment. Other researchers have con-
Additionally, intensive glucose control was not requested firmed this evidence in patients with post-polio syndrome
for enrolled patients. Although strong evidence impli- and Guillain-Barre syndrome [41, 42]. Reducing pro-in-
cates poor glycemic control as a pathogenic mechanism flammatory cytokine levels can downregulate sodium
in the etiology of DN, there is no proof from randomized channels’ expression and induce pain relief. The possibil-
controlled trials that this is the case for pain symptom- ity that IVIG treatment has a normalizing effect on mem-
atology [31, 32] To our knowledge, controlled clinical brane potential, reflecting modulation of Naþ currents,
trials to demonstrate whether intensive diabetes therapy has been speculated to explain the rapid course of clinical
improves extant DPN have not yet been published. improvement after IVIG treatment in some CIDP patients
One noteworthy point was that the placebo effect was [43]. In these patients, the rapid course of clinical im-
less evident than expected. In the inactive arm, there was provement after IVIG treatment would be more sugges-
only a 10% pain reduction, while 30% was expected tive of a change in axonal function (e.g., related to ion
[33]. A possible explanation is that all patients had previ- channel properties) than of structural change. Moreover,
ous ineffective participation in several clinical trials, they all these possible mechanisms of action can explain an ef-
had severe pain unresponsive to therapies, and they had a fect of IVIG on pain that is unusual for an analgesic
negative attitude towards other trials. In this setting, a drug: With onset generally after two to three days, the
possible nocebo effect could be expected. The same evi- maximal effect, achieved by three to five days, can be
dence has been described in a meta-analysis of chronic re- dramatic, including on “soft symptoms” such as sleep,
gional pain syndrome clinical trials [34]. fatigue, general malaise [19].
How can we explain these results? Many researchers We had no significant side effects in our patients, even
have proposed a new direction for the pathogenesis of at the dose of 2 g IVIG/kg. This was probably related to
diabetic neuropathic pain. Multiple preclinical [35] and the administration of a 5% concentration of immunoglo-
clinical [36] studies demonstrate a pathogenic role for bulins over five days, in addition to the good safety profile
inflammation, especially cytokine and chemokine pro- of IVIG, including in patients with neurologic diseases.
duction, in the development of DPN. Accumulated evi- In conclusion, treatment with IVIG was deemed effi-
dence suggests that proinflammatory cytokines and cacious and well tolerated for patients with DPN resis-
chemokines play a part in the pathogenesis of neuro- tant to standard therapies. The present study was
pathic pain. The expression of proinflammatory cyto- designed as a randomized, double-blind, placebo-con-
kines such as IL-1b, TNF-a, and IL-6 (which are trolled clinical trial. It confirms previous data concerning
upregulated in peripheral nerve injury) elicits the inflam- significant pain relief with IVIG in patients with DPN re-
matory process and subsequently elicits neuropathic sistant to standard therapies. Further investigations are
pain [9, 10, 12, 37–39]. necessary to confirm our data. More patients need to be
Overexpression of pro-inflammatory cytokines can enrolled. We hope to involve more centers worldwide for
upregulate sodium channels Nav 1.3 and 1.8. In He a larger clinical trial.
et al., perisciatic administration of recombinant rat TNF-
a (rrTNF) without any nerve injury was shown to pro-
duce lasting mechanical allodynia and to upregulate Acknowledgments
Nav1.3 and Nav1.8 in DRG neurons in vivo, and rrTNF-
Jordi Bozzo PhD, CMPP (Grifols), is acknowledged for med-
a was also shown to enhance the expression of Nav1.3
ical writing assistance and editorial support in the prepara-
and Nav1.8 in cultured adult rat DRG neurons in a dose-
tion of this manuscript, under the direction of the authors.
dependent manner. Furthermore, inhibition of TNF-a
synthesis, which has been demonstrated to prevent neu-
ropathic pain, strongly inhibited the upregulation of References
Nav1.3 and Nav1.8 [12]. Randomized controlled trials 1. Duby JJ, Campbell RK, Setter SM, White JR, Rasmussen KA.
are currently ongoing to investigate further the prelimi- Diabetic neuropathy: An intensive review. Am J Health Syst
nary evidence that IgG may be an effective treatment for Pharm 2004;61:160–73.
Jann et al. 9

2. Tesfaye S, Boulton AJ, Dickenson AH. Mechanisms and man- 21. Jann S, Francia A, Fruguglietti ME, De Toni Franceschini L,
agement of diabetic painful distal symmetrical polyneuropathy. Sterzi R. Efficacy and safety of intravenous immunoglobulin as
Diabetes Care 2013;36(9):2456–65. adjuvant treatment for refractory neuropathic pain. Results of
3. Truini A, De Toni Franceshini L, Zanette G, Praitano M, an open-label, multicenter study. Pain Med 2012;13

Downloaded from https://academic.oup.com/painmedicine/advance-article-abstract/doi/10.1093/pm/pnz331/5697247 by University of Waterloo user on 09 January 2020


Schenone A. Prevalence of peripheral neuropathies and neuro- (10):1334–41.
pathic pain in patients with diabetes. Preliminary results of an 22. Tesfaye S, Boulton AJ, Dyck PJ, et al. Diabetic neuropathies:
Italian multicenter study. J Peripher Nerv Syst 2011;16:S36–7. Update on definitions, diagnostic criteria, estimation of severity,
4. Bouhassira D, Attal N. Diagnosis and assessment of neuropathic and treatments. Diabetes Care 2010;33(10):2285–93.
pain: The saga of clinical tools. Pain 2011;152(Suppl):S74–83. 23. Gould D, Kelly D, Goldstone L, Gammon J. Examining the va-
5. Alleman CJ, Westerhout KY, Hensen M, et al. Humanistic and lidity of pressure ulcer risk assessment scales: Developing and us-
economic burden of painful diabetic peripheral neuropathy in ing illustrated patient simulations to collect the data. J Clin Nurs
Europe: A review of the literature. Diabetes Res Clin Pract 2015; 2001;10(5):697–706.
109(2):215–25. 24. Spallone V, Morganti R, D’Amato C, Greco C, Cacciotti L,
6. Attal N, Cruccu G, Baron R, et al. EFNS guidelines on the phar- Marfia GA. Validation of DN4 as a screening tool for neuro-
macological treatment of neuropathic pain: 2010 revision. Eur J pathic pain in painful diabetic polyneuropathy. Diabet Med
Neurol 2010;17(9):1113–e88. 2012;29(5):578–85.
7. Craner MJ, Klein JP, Renganathan M, Black JA, Waxman SG. 25. Padua L, Briani C, Jann S, et al. Validation of the Italian version
Changes of sodium channel expression in experimental painful of the Neuropathic Pain Symptom Inventory in peripheral ner-
diabetic neuropathy. Ann Neurol 2002;52(6):786–92. vous system diseases. Neurol Sci 2009;30(2):99–106.
8. Hong S, Morrow TJ, Paulson PE, Isom LL, Wiley JW. Early 26. Ware JE, Jr., Sherbourne CD. The MOS 36-item Short-Form
painful diabetic neuropathy is associated with differential Health Survey (SF-36). I. Conceptual framework and item selec-
changes in tetrodotoxin-sensitive and -resistant sodium channels tion. Med Care 1992;30(6):473–83.
in dorsal root ganglion neurons in the rat. J Biol Chem 2004;279 27. Apolone GC, Liberati MC, Mosconi P. Questionario sullo stato
(28):29341–50. di salute SF-36. Traduzione e validazione della versione italiana:
9. Doupis J, Lyons TE, Wu S, Gnardellis C, Dinh T, Veves A. Risultati del progetto IQOLA. Metodol Didattica Clin 1997;5
Microvascular reactivity and inflammatory cytokines in painful :86–94.
and painless peripheral diabetic neuropathy. J Clin Endocrinol 28. Swanenburg J, Gruber C, Brunner F, Wirth B. Patients’ and
Metab 2009;94(6):2157–63. therapists’ perception of change following physiotherapy in an
10. Uceyler N, Rogausch JP, Toyka KV, Sommer C. Differential ex- orthopedic hospital’s outpatient clinic. Physiother Theory Pract
pression of cytokines in painful and painless neuropathies. 2015;31(4):293–8.
Neurology 2007;69(1):42–9. 29. Bouhassira D, Wilhelm S, Schacht A, et al. Neuropathic pain
11. Calvo M, Dawes JM, Bennett DL. The role of the immune sys- phenotyping as a predictor of treatment response in painful dia-
tem in the generation of neuropathic pain. Lancet Neurol 2012; betic neuropathy: Data from the randomized, double-blind,
11(7):629–42. COMBO-DN study. Pain 2014;155(10):2171–9.
12. He XH, Zang Y, Chen X, et al. TNF-alpha contributes to up- 30. Snedecor SJ, Sudharshan L, Cappelleri JC, Sadosky A, Mehta S,
regulation of Nav1.3 and Nav1.8 in DRG neurons following Botteman M. Systematic review and meta-analysis of pharmaco-
motor fiber injury. Pain 2010;151(2):266–79. logical therapies for painful diabetic peripheral neuropathy. Pain
13. Kawasaki Y, Zhang L, Cheng JK, Ji RR. Cytokine mechanisms Pract 2014;14(2):167–84.
of central sensitization: Distinct and overlapping role of 31. Tesfaye S, Vileikyte L, Rayman G, et al. Painful diabetic periph-
interleukin-1beta, interleukin-6, and tumor necrosis factor-alpha eral neuropathy: Consensus recommendations on diagnosis, as-
in regulating synaptic and neuronal activity in the superficial spi- sessment and management. Diabetes Metab Res Rev 2011;27
nal cord. J Neurosci 2008;28(20):5189–94. (7):629–38.
14. Pabreja K, Dua K, Sharma S, Padi SS, Kulkarni SK. Minocycline 32. Ziegler D, Fonseca V. From guideline to patient: A review of re-
attenuates the development of diabetic neuropathic pain: cent recommendations for pharmacotherapy of painful diabetic
Possible anti-inflammatory and anti-oxidant mechanisms. Eur J neuropathy. J Diabetes Complications 2015;29(1):146–56.
Pharmacol 2011 2011;661:15–21. 33. Cepeda MS, Berlin JA, Gao CY, Wiegand F, Wada DR. Placebo
15. Talbot S, Chahmi E, Dias JP, Couture R. Key role for spinal dor- response changes depending on the neuropathic pain syndrome:
sal horn microglial kinin B1 receptor in early diabetic pain neu- Results of a systematic review and meta-analysis. Pain Med
ropathy. J Neuroinflamm 2010;7(1):36–51. 2012;13(4):575–95.
16. Hartung HP. Advances in the understanding of the mechanism 34. Mbizvo GK, Nolan SJ, Nurmikko TJ, Goebel A. Placebo
of action of IVIg. J Neurol 2008;255(Suppl 3):3–6. responses in long-standing complex regional pain syndrome:
17. Wang XL, Dou JT, Zhao H, et al. Chronic inflammatory demye- A systematic review and meta-analysis. J Pain 2015;16
linating polyneuropathy mimicking diabetic neuropathy in a (2):99–115.
young female with type 2 diabetes mellitus. Diabetes Res Clin 35. Cameron NE, Cotter MA. Pro-inflammatory mechanisms in dia-
Pract 2012;96:e15–7. betic neuropathy: Focus on the nuclear factor kappa B pathway.
18. Boukhris S, Magy L, Khalil M, Sindou P, Vallat JM. Pain as the Curr Drug Targets 2008;9(1):60–7.
presenting symptom of chronic inflammatory demyelinating 36. Hur J, Sullivan KA, Pande M, et al. The identification of gene ex-
olyradiculoneuropathy (CIDP). J Neurol Sci 2007;254(1– pression profiles associated with progression of human diabetic
2):33–8. neuropathy. Brain 2011;134(11):3222–35.
19. Goebel A, Blaes F. Complex regional pain syndrome, prototype 37. Skundric DS, Lisak RP. Role of neuropoietic cytokines in devel-
of a novel kind of autoimmune disease. Autoimmun Rev 2013; opment and progression of diabetic polyneuropathy: From glu-
12(6):682–6. cose metabolism to neurodegeneration. Exp Diabesity Res 2003;
20. Tamburin S, Borg K, Caro XJ, et al. Immunoglobulin g for the 4(4):303–12.
treatment of chronic pain: Report of an expert workshop. Pain 38. Younger DS, Rosoklija G, Hays AP, Trojaborg W, Latov N.
Med 2014;15(7):1072–82. Diabetic peripheral neuropathy: A clinicopathologic and
10 IVIG for Painful Diabetic Neuropathy

immunohistochemical analysis of sural nerve biopsies. Muscle 41. Gonzalez H, Khademi M, Andersson M, et al. Prior
Nerve 1996;19:722–7. poliomyelitis-IVIg treatment reduces proinflammatory cytokine
39. Catanzaro O, Capponi JA, Michieli J, Labal E, Di Martino I, production. J Neuroimmunol 2004;150(1–2):139–44.
Sirois P. Bradykinin B(1) antagonism inhibits 42. Reuben S, Sumi MG, Mathai A, Nair MD, Radhakrishnan VV.

Downloaded from https://academic.oup.com/painmedicine/advance-article-abstract/doi/10.1093/pm/pnz331/5697247 by University of Waterloo user on 09 January 2020


oxidative stress and restores NaþKþ ATPase activity in dia- Intravenous immunoglobulin reduces serum tumor necrosis fac-
betic rat peripheral nervous system. Peptides 2013;44 tor alpha in patients with Guillain-Barre syndrome. Neurol India
:100–4. 2003;51(4):487–9.
40. Sharief MK, Ingram DA, Swash M, Thompson EJ. I.v. immu- 43. Lin CS, Krishnan AV, Park SB, Kiernan MC. Modulatory effects
noglobulin reduces circulating proinflammatory on axonal function after intravenous immunoglobulin therapy in
cytokines in Guillain-Barre syndrome. Neurology 1999;52 chronic inflammatory demyelinating polyneuropathy. Arch
(9):1833–8. Neurol 2011;68(7):862–9.

You might also like