On the dark side of therapies with immunoglobulin concentrates: the adverse events

38
On the dark side of therapies with immunoglobulin concentrates. The adverse events Peter J. Spaeth, Guido Granata, Fabiola La_Marra, Taco W. Kuijpers and Isabella Quinti Journal Name: Frontiers in Immunology ISSN: 1664-3224 Article type: Review Article First received on: 31 Oct 2014 Frontiers website link: www.frontiersin.org Primary Immunodeficiencies

Transcript of On the dark side of therapies with immunoglobulin concentrates: the adverse events

   

 On the dark side of therapies with immunoglobulin concentrates. Theadverse events

  Peter J. Spaeth, Guido Granata, Fabiola La_Marra, Taco W. Kuijpers and Isabella Quinti

Journal Name: Frontiers in Immunology

ISSN: 1664-3224

Article type: Review Article

First received on: 31 Oct 2014

Frontiers website link: www.frontiersin.org

Primary Immunodeficiencies

1

Fact Sheet 1 2

Tile of the manuscript: On the dark side of therapies with immunoglobulin 3

concentrates. The adverse events. 4 5 Submission to: Frontiers in Immunology, section Primary Immunodeficiencies, special issue on 6

“Immunoglobulin therapy in the 21st century: the dark side of the moon” 7 8 Article type: review 9 10 Number of authors: 5 11 12 Number of institutions: 3 13 14 Character counting of the abstract, spaces included: 1919 (2000 allowed) 15 16 Word counting body text: 5905 (12000 allowed) 17 18 Word counting acknowledgments: 24 19 20 Word counting legend to figures: 944 21 22 Number of tables: 3 23 24 Number of figures: 8 25 26 Number of references cited: 178 27

28

2

On the dark side of therapies with immunoglobulin concentrates. 29

The adverse events 30

31 Peter J. Späth1, Guido Granata2, Fabiola La Marra2, Taco W. Kuijpers3 and Isabella Quinti2* 32 33 1Institute of Pharmacology, University of Berne, Berne, Switzerland, 2Dept.of Molecular Medicine, 34 Sapienza University of Rome, Rome, Italy and 3Department of Pediatric Hematology, Immunology 35 & Infectious disease, Academic Medical Centre (AMC), University of Amsterdam, Amsterdam, 36 Netherlands 37 38 39 Conflict of interest statement 40 The authors declare a potential conflict of interest and state it below. 41 PJS, none; GG, none; FLM, none; TWK, none; IQ, none 42 43 44 Running Title: Adverse events of Ig therapies 45 46 47 Correspondence 48 Isabella Quinti, MD, PhD 49 Dept.of Molecular Medicine, Sapienza University of Rome 50 Viale dell’Universita 37 51 I-00186 Rome 52 Italy 53 54 [email protected] 55 56 tel. and fax. +390649972007 57

58

3

Abstract to the dark side of therapies with human immunoglobulin G concentrates 59 60 Therapy and manufacturing of human immunoglobulin G (IgG) concentrates is a success story 61 ongoing for decades showing an ever increasing demand for this plasma product. Transmission of 62 pathogens in the last decade could be entirely controlled through the antecedent introduction by 63 authorities of a regulatory network and installing quality standards by the plasma fractionation 64 industry. A cornerstone of the regulatory framework is current Good Manufacturing Practice 65 ensuring by validated steps the constant quality of an IgG concentrate. One of the more remarkable 66 steps to achieve pathogen safety was the introduction of virus filtration. Virus filtration became a 67 very versatile tool to eliminate e.g. the suspected agent of variant Creutzfeldt-Jakob disease. The 68 success of IgG concentrates on a clinical level is documented by the slowly increasing number of 69 registered indication and the more rapid increase of the off-label uses. A part of the success is the 70 adverse event profile of IgG concentrates which, even at life-long need for therapy, is excellent. 71 Adverse events (AEs) occur rarely and mainly are mild to moderate. Below we review AEs, give 72 insight in to possible mechanisms, and particularly focus on hemolysis and thrombosis. A further 73 interest of this review is the question how the introduction of plasma fractionation by ion-exchange 74 chromatography and polishing by immunoaffinity chromatographic steps might influence AE 75 profiles and efficacy of IgG concentrates. Indeed, it is not clear whether the improved recovery, 76 which can be achieved by the ion-exchange chromatography technique, leads to the expansion of 77 the same repertoire of IgG molecules in a product or whether some IgG specificities might be lost 78 while specificities otherwise absent or present in minute amounts only contribute to the improved 79 recovery. Latter might have consequences on the AE profile of a new IgG concentrate.80

4

81 1. The tinge of the dark – Introduction clinical manifestations of AEs 82 83 Since the initial clinical use of immunoglobulin G (IgG) concentrates of human origin, transmission 84 of pathogens and non-infectious adverse events (AEs) were reported [1-7]. Before the mid 90ies, 85 transmission of pathogens has depended on the pool size and the fractionation methods used, 86 particularly the polishing steps of an IgG concentrate [8]. Mode of fractionation, i.e. cold ethanol or 87 ion-exchange chromatography, contaminants, route of application, i.e. intra muscular (IMIG), 88 intravenous (IVIG) or subcutaneous (SCIG), the rate of increase of the exogenous IgG in the 89 circulation of the recipient over time (Fig. 1) and incorrect handling of the concentrate are factors 90 having a role in inducing non-infectious AEs related to administration of IgG concentrates (Table 91 1). IgG concentrates represent a defined part of the adaptive immune system, are isolated form 92 pooled human plasma of at least 1000 donors, which contribute to the diversity in the final plasma-93 derived product. Therapies with IgG concentrates manufactured according to regulators 94 requirements are acknowledged to be safe in general. This does not exclude the occurrence of AEs 95 which in in their majority are rare and clinically mild to moderate. Below we like to give a few 96 insights into various aspects and possible mechanisms of AEs. 97 98 99 2. A dark and frightening environment – Pathogen safety of IgG concentrates 100 101 Modern IgG concentrates from the moment of sampling whole blood and isolation of plasma 102 thereof (recovered plasma) or the machine-supported collection of plasma (source plasma, apheresis 103 plasma) has to occur in a regulatory framework and the quality standards implemented by the 104 plasma fractionating industry (Fig. 2). The cornerstone of the regulatory framework is current Good 105 Manufacturing Practice (cGMP). The pillars of pathogen safety are validated virus inactivation and 106 virus elimination steps (Fig. 3). A hallmark of virus elimination introduced in the late 90ies in 107 Berne by the team of Christoph Kempf is the large scale virus filtration technique (formerly also 108 termed ‘nanofiltration’) [8]. Meanwhile, virus filtration became a very versatile tool to eliminate a 109 variety of pathogens, e.g. the suspected agent of variant Creutzfeldt-Jakob disease. Thanks to the 110 tightly implemented regulatory framework pathogen safety of plasma products is at a level never 111 reached before. This is well supported by the fact of missing reports of transmission by IgG 112 concentrates of emerging viruses (SARS coronavirus, West Nile Virus, MERS coronavirus and 113 others), zoonotic pathogens or the agent of variant Creutzfeldt-Jacob disease in the last decade 114 [9,10] and development of potentially specific screening techniques to eradicate transmission of 115 vCJD in the future[11]. 116 117 118 3. Some basic insights into possible mechanisms occurring in the dark 119 120 The human immune system is in charge of controlling invading organisms and provides peripheral 121 and tissue homeostasis by a variable-region connected network. The latter supports the efficient 122 removal of roughly ~1012 altered/senescent cells of the body per day. Host defense is best mediated 123 by ‘immune antibodies’, which have undergone somatic hypermutations and have narrow 124 specificities and high affinities. Antibodies, including the homeostatic ones, produced in absence of 125 an antigenic stimulation are termed ‘natural antibodies’ (NAbs). They are of germ-line 126 configuration or have undergone a few somatic hypermutations only. In general they have broad 127 specificity, are of low affinity and high avidity (with exceptions) and participate in primary host 128 defense (i.e. react best with repetitive structures as can be found on bacteria). Their homeostatic 129 functions are brought about by self-reactive antibodies which we like to term physiologic 130 autoantibodies. The immunoglobulins pools in mammals (IgM, IgG and IgA) to its smaller part 131

5

provides defense and to its larger part homeostasis. Human IgG has a role in both. Thus, 132 infusion/injection of IgG concentrates inevitably results in recognition of occasional pathogens, 133 toxins or superantigens and concomitantly in a wide array of variable-region (V-region) 134 interactions. V-region recognitions are interactions of the self-reactive antibodies of the many 135 donors in the IgG concentrate with the recipients’ body/immune system and vice versa. A 136 bewildering wide range of possible reactions can occur which primarily are dependent on the 137 immunologic status of the recipient at the time of therapy and to a smaller part on the IgG 138 concentrate(s). The therapeutic effect achieved depends on the disease treated, and can depend on 139 the concentration reached locally, i.e. can have agonistic or antagonistic effects [11,12]. In 140 summary, it is our opinion that IgG concentrates always provide more or less the same ‘bouquet’ of 141 IgG specificities (similarity). Each time, the recipient’s actual immune condition decides from 142 which part of the IgG the patient’s derailed immune system is profiting (diversity). 143 144 Parameters leading to adverse events (AEs) related to infusion/injection of IgG concentrates are: (i) 145 the content in the product of biologically highly active likely beneficial ingredient that have to be 146 kept under control (e.g. content of ‘dimers’ devoid of remarkable complement activation in vivo; 147 see below), and the content in unwanted active ingredient that have to be discarded during 148 manufacturing (alloantibodies); (ii) impurities such as IgA (anaphylactoid reaction); (iii) activated 149 coagulation and contact activation factors (thromboembolic events) and; (iv) excipients such as 150 sucrose (osmotic nephrosis). Here, we like to add an additional parameter: the sum of potentially 151 beneficial V-region connected reactions. In case they overshoot they might add to processes leading 152 to AEs. In summary, the intensity of the resulting AEs is depending on the immune status of the 153 recipient, the infusion rate, e.g. how rapidly the active ingredients (the various IgG specificities), 154 the impurities and the excipients reach the circulation of the recipient. Thus, the i.v. application has 155 the highest chance for the occurrence of adverse events. 156 157 In the early days of IVIG therapy, complement-mediated ‘anaphylactoid’ (i.e. immediate) and 158 ‘phlogistic’ (i.e. inflammatory) AEs were distinguished [13,14]. The complement-mediated AEs 159 were considered to be caused by aggregates in the product (‘spontaneous complement activation’ or 160 anti-complementary activity or ACA) or by in vivo formation of immune complexes (patient’s 161 condition related; e.g. subclinical infections or the unnoticed presence of anti-IgA antibodies) and 162 therefore only IgG concentrates with low or absent ACA is accepted by authorities for human use. 163 Below, we present one instructive case of each type of reaction. 164 165 166 3.1. The rapid onset of darkness – The immediate adverse events 167 168 The first reports of AEs concerned either the application of complement-activating fractions in an 169 IgG concentrate or the in vivo formation of complement-activating immune complexes [2-4]. A 170 very rare but potentially fatal condition is the formation of IgA/anti-IgA complexes in patients 171 being initiated on replacement therapy and having serum IgG antibodies against infused IgA not 172 recognized before the start of the IVIG infusion [15]. Prerequisite for the presence of anti-IgA 173 antibodies is the most common primary immunoglobulin defect, i.e. selective IgA deficiency 174 (sIgAD) or IgAD associated with diminution of other immunoglobulin classes. IgA deficiency is 175 defined by serum levels of <0.05 g/L or <0.07 g/L (depending on laboratories). A marked 176 diminution of serum IgA consistent with IgAD in various ethnic groups is estimated being 1:155 to 177 1:18,550 [16]. The mean frequency in Caucasians is approximately 1:700 [17]. Up to 40% of 178 patients with IgAD have been reported having anti-IgA antibodies in the serum with titers ranging 179 between 1:4 and 1:262,144. In approximately 10% of patients with common variable 180 immunodeficiency, and occasionally in patients with other primary immunodeficiency diseases 181 measurable anti-IgA can be detected [18,19]. These antibodies are predominantly of the IgG class, 182

6

but anti-IgA antibodies of other immunoglobulin classes have been described as well [20,21]. The 183 reason for their emergence remains unknown. 184 185 Taken the above numbers the infusion of human derived products containing IgA resulting in severe 186 anaphylactoid type AEs should be considerable. This is not the case [22]. Questions about the 187 clinical relevance of above numbers emerge as soon as (i) blood banks estimate the theoretical risk 188 of IgA anapyhlactic reactions [15]; (ii) assess relation of severe IgAD with presence or absence of 189 anti-IgA antibodies [23]; (iii) screen donors for very low IgA levels in order to become able to 190 provide blood and plasma derived products free of IgA and find a considerably lower frequency 191 than expected [24]; (ii) when a more close look to ‘anti-IgA’ gives ‘unexpected’ results, including 192 ‘anti-IgA’ in blood donors with normal serum IgA level or ‘anti-IgA’ that cannot be neutralized 193 with purified IgA [25]; (iv) or when blood products containing proven anti-IgA do not elicit severe 194 AEs [26]. 195 196 Among patients on replacement therapy those with CVID may rarely develop severe AEs [15]. The 197 discrepancy between anti-IgA positive patients and frequency of AEs raises the question about the 198 nature of the many reported anti-IgA antibodies and also raises the question about the immunologic 199 condition which allows the formation of anaphylactoid anti-IgA antibodies. There might be some 200 logic in supposing, anaphylactoid anti-IgA cannot evolve at IgA levels otherwise fulfilling the 201 definition of IgAD. Such a condition would constantly generate immune complexes which in turn 202 could activate complement, react with immune cells and be deposited in lung and kidney. Indeed, 203 Horn et al. found anti-IgA antibodies in CVID patients missing IgA+ B cells and presenting with 204 IgA levels <0.0009 g/L, a level which is more than 50 to 70 fold lower than the threshold for IgAD 205 [27]. 206 207 The kinetics of anti-IgA after infusion of blood products have been studied in a few cases. In these 208 patients, a fall in anti-IgA titers has been noticed followed by an increase during subsequent weeks 209 or months. This suggests that at appropriate proportions, IgA of the infused material and anti-IgA 210 present in the patients’ serum combine with each other to form immune complexes, activate 211 complement, and are eliminated by macrophages. The increase in anti-IgA titers over time indicates 212 that the infused IgA-containing product has a booster effect [19,20,28]. Such boosting effect 213 together with the presence of anti-IgA before the application of an IgG concentrate can be taken as 214 the ultimate confirmation of a supposed IgA/anti-IgA (Fig. 4). 215 216 A non-complement-mediated anaphylactoid reaction was ascribed to the unforeseen release of 217 elastase and other pro-inflammatory substances from neutrophils activated by the formation of in 218 vivo IgA/anti-IgA complexes. Complement-activation or mast cell-dependent release of vasoactive 219 substances were excluded as pathogenic mechanisms. Although the IgA/anti-IgA complexes usually 220 do not cause clinically relevant neutrophil degranulation within the circulation, the presence of a 221 rare genotype encoding a novel gain-of-function IgG receptor on neutrophils may provoke 222 premature degranulation by these complexes. This phenomenon was only relevant in 223 hypogammaglobulinemic patients in the presence of in vivo IgA/anti-IgA complexes [29]. The low 224 prevalence of this genotype combined with an IgAD or CVID may explain the rarity of serious 225 anaphylactoid reactions in newly IVIG-treated patients. 226 227 228 3.2. Insight into the phlogiston of the dark – The pro-inflammatory cytokines 229 230 In the early days of Ig-therapy the nature of the ‘phlogistic’ AEs was obscure. However, it was 231 already known that initial sings of AE can be overcome when the patient receives a dose of IVIG 232 his immune system can cope with, than infusion is stopped for several hours, where after infusion 233

7

can be restarted at high rates without further problems (Fig. 5). One of the authors had a particular 234 opportunity to get an insight into what a “phlogistic reaction” might be. At the occasion of a 235 voluntary infusion of an investigational liquid IVIG, he encountered a severe flu-like AE of more 236 than 12 hours duration. Before injection, the investigational liquid preparation had passed all release 237 criteria for human use, including spontaneous complement activation assessed by ACA and was 238 free of prekallikrein activator. In those days assays for cytokines in biological samples just began to 239 become available and were included into the parameters assessed in the study. Infusion was stopped 240 after one hour because of a drop of pulse rate and heavy discomfort provoking the laconic comment 241 by the proband’s chief technician who was taking samples: “you look green”. The infusion was 242 continued after another 90 minutes when the heart rate had almost normalized. The infusion could 243 be completed within an additional 3.25 hours (0.4g/kg b.w.) without further aggravation of malaise. 244 The leukocyte count transiently had dropped to a nadir of 40% at 2 hours followed by a 245 leukocytosis peaks at 8 hours. Complement activation, as assessed by generation of 246 C3a/C3a[desArg] and the formation of the terminal complement complex C5b-9, apparently did not 247 occur: the C3a/C3a[desArg] value reached a maximum of 260 ng/mL (norm: <200 ng/mL) at 7 248 hours while the C5b-9 value never moved outside the normal range. Instead, a sequence of rapid 249 transient massive increases of pro-inflammatory cytokines was seen: (i) TNF� started to increase 250 30 min post initiation of infusion from a value of 20 pg/mL to a peak value at two hours which was 251 above the calibration range of the test kit of 1500 pg/mL; (ii) IL-8 increase started after 1 hour from 252 29 pg/mL and peaked at 2.5 hours with 4400 pg/mL post initiation of infusion; (iii) IL-6 secretion 253 started after 1 hour with an undetectable level and peaked at 4 hours with 345 pg/mL. All pro-254 inflammatory cytokines fell sharply while the second part of the infusion was still ongoing. The day 255 after infusion, the pro-inflammatory cytokine profiles were back to normal and the flu-like 256 syndrome was gone. In contrast, IL-1ra values started increasing at 1.5 hours (300pg/mL), peaked at 257 4 hours (>32000 pg/mL) and decreased slowly to reach a value of 10500 pg/mL 24 hours after 258 initiation of the infusion. 259 260 A series of further experiments with investigational and marketed IVIGs was performed. All IgG 261 concentrates were analyzed for their molecular weight (MW) distribution. The most remarkable 262 differences emerged in the MW range of dimers while the presence of minute amounts of higher 263 oligomers could not be excluded with certainty. Below we will use the term ‘dimers’ for that 264 fraction of IgG with higher MW. Subsequent findings indicated that levels of ‘dimers’ >12% were 265 responsible for complement-independent cell activation and cytokine release. The TNF peaks 266 assessed at 2.5 hours post initiation of infusions correlated with ’dimer’ content of the IVIGs and 267 mirrored a clinical score of AEs [30-32]. 268 269 A few years before a complement-independent induction of a hypotensive factor by IgG di- and 270 oligomers was reported in animal experiments [33]. A key role for macrophages in the generation of 271 the hypotensive lipid factor was identified as platelet-activating factor, being induced by dimers and 272 polymers [34,35]. Several years later, the dimer-mediated AEs in animal experiments were 273 confirmed [36,37]. Yet, at the same time, the dimer content of IVIG apparently correlated with the 274 clinical efficacy in a murine ITP model [38,39]. Variables such as ‘infusion rate’, ‘genetic 275 background’, ‘endogenous immunoglobulin levels’, or ‘proportional fraction of polymers versus 276 dimers’ may impact on the balance between the phlogiston (being cytokines, active lipid 277 substances, or a combination of factors) and the therapeutic efficacy (blocking IgG receptors on 278 liver/spleen macrophages to prevent clearance of ‘opsonized’ material such as platelets in ITP). As 279 of today, reports on release of cytokines in humans in association with AEs or tolerability toward 280 dimers remain scarce and to the best of our knowledge studies in humans of causative 281 factors/fraction in an IgG concentrate has not been adequately addressed [40-43]. 282 283

8

In IgG preparations, various forms of dimers might be present: formed through covalent binding 284 [44] by denaturation, hydrophobic interactions of the Fc-parts and by idiotype/anti-idiotype 285 interactions, as part of the V-connected network of peripheral immune homeostasis [45]. For a 286 commercially viable fractionation process, pooling of donated plasma is mandatory in order to 287 obtain a volume of starting material large enough to cover ever increasing costs for documentation, 288 in-process and batch-release testing as it is required by cGMP. Pooling also intends to smoothing 289 the batch-to-batch differences in antibody titres, a goal apparently difficult to achieve to levels as 290 theory might imply [46]. Consequences of pooling are on the one hand an enrichment of 291 public/common immune antibodies while diluting out individual specificities; on the other hand, the 292 antibodies of the immune network of an individual donor are exposed to those of many other 293 donors. The more individuals contribute to the pool, the more complex the possible ‘immune-294 network’ interactions will become. The subsequent fractionation process has far-reaching effects on 295 immunoglobulins from a given pool: only trace amounts of IgA and IgM are retained in the final 296 product, i.e. IgG is deprived of its counterparts of the V-connected immune network. The ‘naked’ 297 IgG molecules of the homeostatic network can interact with each other at a ’random combining site-298 interactions of single donor-derived monomeric IgG’ [47], otherwise not existing in vivo. This 299 interaction is largely reversible. With increasing numbers of donors included into the pool, the 300 immune network recognition among the ‘naked’ IgG molecules of the V-connected network 301 becomes more and more complex, and the dimer and lower oligomer content in the resulting IgG 302 concentrate increases [48-50]. In lyophilized IgG concentrates, the dimer formation is ‘frozen’ at a 303 low level while in liquid preparations an equilibrium between monomers and dimers is achieved 304 over time reaching a dimer content of 12% or more if not hampered by stabilizers. Specificities, as 305 far as they have been addressed, in the dimer fraction considerably differ from the monomeric 306 fraction [51-55]. In conclusion, the immunomodulatory efficacy of IgG concentrates in part depends 307 on the capacity and extent to form ‘dimer’ fractions devoid of remarkable complement activation in 308 vivo. The ‘art’ of manufacturing a liquid IgG concentrate is not to eliminate the monomeric IgG 309 having potential for ‘dimer’ formation but to inhibit extensive ‘dimerization’. In summary, AEs 310 might be associated with the induction of pro-inflammatory cytokines in absence of measurable 311 complement activation in vivo where all regulatory mechanisms and removal processes of a body 312 are at disposition. At reasonable infusion rates in the open system of the human body, clinically 313 relevant systemic complement activation apparently needs oligomers formed of three or more IgG 314 molecules. 315 316 317 4. The missing oxygen on the dark side - Immunoglobulin-induced hemolysis 318 319 There are multiple reports of Ig-induced hemolytic anemia (HA) in patients receiving high doses of 320 IVIG [56-91] (Table 2 and Fig.6; (www.adrreports.eu). By spontaneous reporting, risk factors 321 recognized for Ig-induced hemolysis include beside high doses (more than 100 g IVIG over 2-4 322 days), female gender and histo-blood group type A, B or AB of recipients. 323 324 A significant proportion of patients receiving IVIG develop a positive direct antiglobulin test 325 (DAT) detectable after 24 hours for up to 10 days after the IVIG infusion [92,93]. However, it 326 should be underlined that the DAT positivity due to the factors mentioned above [94-95] is not 327 sufficient per se to diagnose hemolysis and DAT positivity does not necessarily imply the presence 328 of active hemolysis. DAT-positive mild hemolytic reactions can be easily missed and the true 329 incidence of such reactions is difficult to document without careful clinical and laboratory follow-330 up. 331 332

9

In the majority of reports on HA, intravascular RBC destruction via complement activation or 333 extravascular RBC sequestration and removal by the reticulo-endothelial system was proposed to 334 result from IgG alloantibodies with specificity for RBC antigens A, B, D or C. 335 HA induced by high-dose IVIG has an average incidence of 5.8% [96]. Low dose IgG replacement 336 therapy is considered universally as safe, and only few cases of hemolysis following low-dose IVIG 337 SCIG administration have been described [63,78,93]. A baseline white and red blood cell count 338 prior to IVIG initiation and a close clinical and laboratory follow up was suggested as a useful tool 339 for early diagnosis and treatment. A possible work up might be to check hemoglobin (Hb) level 340 prior and 48-78 hours after Ig infusion. In case of a drop of Hb, the presence of DAT, an increase in 341 unconjugated bilirubin, lactate dehydrogenase (LDH), and reduced haptoglobin level, followed by a 342 rise in reticulocyte count should be assessed (Fig.7). We systematically reviewed case reports 343 related to IVIG-induced hemolysis from 1987 to 2014 and identified 29 articles containing reports 344 of 109 patients. Baseline characteristics of the patients are shown in Table 2. When available, blood 345 group, DAT, Hb drop and outcome are indicated. All reports showed positive DAT, except for a 346 case of Yin et al [59]; in this case, DAT was performed 10 days after IVIG administration and the 347 DAT negativity might have been due to a rapid removal of sensitized RBCs. 348 349 In the majority of patients, the outcome was positive: 106 out of 109 patients recovered with or 350 without packed RBC transfusions; three patients died after HA, with the hemolytic episode 351 representing a precipitating factor of a severe underlying condition. Elution experiments were 352 performed and the search for blood group antibodies revealed anti-A and anti-B specificity in the 353 majority of cases; anti-D specificity was assessed in 4 reports, often associated with other 354 specificities [78,89,93]. A search for other specificities such as anti-band 3 or anti-Gal was not 355 performed. Only one report detected anti-C specificity in three patients; in one of them associated 356 with anti-D irregular antibodies [93]. Although studies were restricted to blood group antibodies, 357 this finding demonstrated that polyvalent IgG preparations might contain clinically significant non-358 blood group antibodies, which are not part of the lot-release criteria in that their titration is not yet 359 required by the European Pharmacopoeia. Antibodies in HA such as anti-C may have unexpected 360 hemolytic consequences [97-101]. Beside passive transfer of alloantibodies, IgG administration 361 also has been demonstrated to lead to unspecific enhanced erythrocyte sequestration, in particular in 362 patients with underlying inflammatory disorders [102-103]. In 2009, the Canadian IVIG hemolysis 363 Pharmacovigilance group elaborated criteria to define an ‘IVIG-induced hemolysis’ [66]. They 364 included a reduction of Hb levels ≥ 1 g within 10 days after Ig administration, with appearance of a 365 positive DAT and, at least, two of the following criteria: increase in the reticulocyte count, elevation 366 of lactate dehydrogenase (LDH) and unconjugated bilirubin serum levels, low haptoglobin, 367 hemoglobinuria, hemoglobinemia, presence of significant spherocytosis, in the absence of 368 alternative causes of anemia. The passive transfer of IgG alloantibodies through IgG concentrates is 369 difficult to explain as polyvalent IgG is prepared from plasma of thousands of donors. Since 370 immunization to RBC alloantigens can occur because of past transfusions or pregnancy, the 371 hypothetical numbers of alloimmunized plasma donors should be rather low. Recently, other 372 mechanisms underlying alloimmunization related to molecular mimicry have been demonstrated 373 [104]. The mechanism of high-dose IVIG-induced HA is complex and it might vary from patient to 374 patient. IVIG cause hemolysis due to: (i) disease-associated pre-coating of RBCs; (ii) IgG with 375 hemolysis triggered by passive transfer of IgG binding to blood group antigens; (ii) transfer of high 376 levels of alloantibodies to RBC pre-coated at a low level only; or (iv) transfer of clinically tolerable 377 levels of isoagglutinins plus transfer of additional RBC-reacting physiological autoantibodies. 378 Indeed, hemolytic reactions could not be related exclusively to transfer of alloantibodies. Hence, 379 antibodies other than histo-blood group alloantibodies (pre-)coated to RBCs might contribute to 380 hemolysis in IgG recipients need to be identified. In addition, hemolytic episodes may possibly be 381 precipitated by some sort of complexed/denatured IgG that co-purify with other IgG in the product 382 [58,102,103,105]. Recently, a two hit mechanism for IVIG-induced hemolysis has been proposed: 383

10

the passive transfer of alloantibodies through IVIG representing the first hit and the underlying 384 inflammatory state representing the second hit. [106]. Nowadays, all commercial Ig products have 385 to undergo anti-A and anti-B testing and regulatory requirement ask for respective IgG antibody 386 titers of ≤1:64 at 5% solution strength (w/v) [86,87]. Nevertheless, hemolysis might occur even in 387 recipients of IgG products that meet these specifications [58]. Consequently, it has been suggested 388 that IgG recipients should be monitored for clinical signs and symptoms of hemolysis [107]. 389 390 391 4.1. Reasoning in the dark about the consequences of the struggle to stay on the sunny side – 392 Measures to overcome elevated frequencies of hemolysis. 393 394 With the detection of the immunomodulatory potential of IgG concentrates, their clinical use has 395 continuously increased [108]. To cover the need, at a first glance, an increase of the volume of 396 plasma fractionated seems to be the most convenient option. However, this might economically not 397 be viable because plasma products are interconnected [109] and before increasing output of one 398 product (e.g. IVIG) the market absorbance of the other products as well (e.g. albumin) must be 399 ascertained. On a longer-run, a more viable option is to improve recovery. Considering recovery, 400 the cold ethanol fractionation apparently has reached its limits. As of today, four manufactures have 401 invested into a ‘modern’ fractioning technique on the basis of ion-exchange chromatography. Ion-402 exchange chromatography allows elevated recovery at high purity. As of today, five IVIGs, one 403 SCIG and one anti-D concentrate are fractionated by ion-exchange chromatography. 404 Pharmacovigilance has shown that all of them, more or less prominently, show a tendency for 405 elevated frequencies of hemolytic AEs. Anti-A and anti-B alloantibody titers are now lot release 406 criteria (see above) as they constitute the major risk parameter for hemolytic reactions mediated by 407 IgG concentrates. To stay on the sunny side, two manufacturers have taken measures to reduce anti-408 A and anti–B titers in their IgG products. One measure chosen was adsorption of the alloantibodies 409 by affinity chromatography [110]. Reported reduction in both alloantibodies was significant and 410 levels were similar to those in cold-ethanol fractionated immunoglobulins [111]. The other measure 411 chosen was reduction in anti-A using an automated indirect agglutination test for donor screening 412 and exclusion of high-titre donations (approx. 5.1%) from plasma pooling and fractionation [112]. 413 This measure reduced anti-A in the IgG concentrate by one titer step. To ensure staying on the safe 414 and sunny side, the manufacturer has announced the introduction of an alloanti-A and alloanti-B 415 immune-affinity chromatography step into the manufacturing process [113]. Preliminary results 416 indicate depletion in anti-A and anti-B by >80% in investigational lots. Subsequently we want to 417 discuss possible consequences of (extensive) removal of antibodies reacting with histo-blood group 418 antigens A and B. 419 420 421 4.1.1. Struggling to stay on the sunny side – Reduction of histo-blood group A and B 422 alloantibodies in IgG concentrates 423 424 Three facts have initiated our thinking about possible consequences of removal of histo-blood group 425 A and B reacting antibodies from IgG concentrates. (I) in collaboration with Hans U. Lutz, formerly 426 Biochemistry ETH Zurich, we have observed the non-intended removal of natural anti-C3 427 autoantibodies regulating complement activation by large-scale immune-affinity adsorption of IgA 428 from an IgG concentrate [114]. Anti-C3 antibodies belong to the family of ‘natural antibodies’ 429 (NAbs) and have a particular role in homeostasis: they control activation of complement, among 430 others, in the frame of NAb-mediated opsono-phagocytosis of altered or senescent cells, including 431 RBCs [115-117]. Thus, the intention to target one particular antibody by affinity chromatography 432 might reduce that antibody specificity but at the same time affect other specificities as well. (II) it 433 should be kept in mind that the blood groups A and B are in fact ‘histo-blood group’ antigens, i.e. 434

11

they are also found on white blood cells, T lymphocytes and proteins and also can be found in 435 soluble form [118]. Alloantibodies reacting with histo-blood group antigens A and B thus have 436 much broader tissue recognition than RBCs only. In addition, alloanti-A and alloanti-B belong to 437 the population of NAbs recognizing non-self and most likely participate in primary host defense 438 [119]. (III) in contrast to cold-ethanol fractionation, where low titers of alloanti-A and alloanti-B 439 are achieved on basis of their isoelectric points (IEPs), the (extensive) immune affinity removal 440 might affect a much wider IEP range, thereby removing broadly reacting antibodies and impairing 441 some desirable functions of the IgG concentrate. 442 443 444 4.1.2. Possible consequences of the struggle to stay on the sunny side - Antibodies reacting 445 with the terminal di- tri- and tetra-saccharides of histo-blood group antigens A and B 446 447 Histo-blood group A and B epitopes in terminal position are tetrasaccharides. Alloantibodies to 448 these tetra-saccharides are found in plasma of healthy individuals depending on the blood group 449 they have. Research of antibodies other than the alloantibodies reacting with ‘blood group’ A or B 450 has been performed using the corresponding terminal di- or tetra-saccharides for isolation [120-451 122]. The antibodies reacting with terminal di-, tri- and tetra-saccharides of histo-blood groups A 452 and B belong to the large family of human natural anti-glycan immunoglobulins (anti-glycan 453 NAbs). Recently, the repertoire and epitope specificity of such immunoglobulins was addressed in 454 depth [123,124]. It proved that serum of healthy individuals contain respectable amounts of di- or 455 tri-saccharide-reacting NAbs. These pseudo-anti-A and pseudo-anti-B NAbs are not reacting with 456 the tetra-saccharide of histo-blood groups A and B. In contrast alloanti-A and –B antibodies able to 457 react with tetrasaccharides are reacting with corresponding terminal di- and tri-saccharides as well. 458 Understanding the biological role of these ‘high-titer and population conservative’ anti-glycan 459 NAbs needs further investigations and those reacting with histo-blood group A- and B-like 460 structures are of particular interest. A population of the anti-glycan NAbs are the anti-αGal NAbs 461 which recognize Galα1-3Gal and Galα1-3(Fucα1-2)Gal epitopes. Anti-αGal NAbs have been 462 described being xenoreactive, recognizing bacterial Galα1-3Gal [125] and having tissue 463 homeostatic function. The daily removal of altered/senescent cells of the body is ~1012. Removal is 464 mainly mediated by apoptosis (no inflammation, no necrosis). Removal of RBCs with a daily 465 turnover of ~2 x 1011, corresponding to ~20 g cell mass, is effectuated in absence of apoptosis 466 because of the missing nuclei. Instead, in parallel of their age, RBC encounter increasing 467 cumulative oxidative stress, which causes increased exposure of otherwise cryptic structures such as 468 spectrin, band 3 or αGal epitopes. These exposed structures are recognized by low affinity, high 469 avidity NAbs, which promote their efficient removal of RBCs [126]. Immunoaffinity adsorption of 470 Gal by trisaccharides might have a Janus effect. Directing to the sunny side of the moon, 471 adsorbing Gal NAbs reacting with altered and senescent self on RBC might prevent an increase in 472 the IgG load of RBCs of individuals with inflammatory diseases eventually facilitating hemolysis. 473 Directing to the dark side of the moon, such adsorption of tissue homeostatic antibodies might 474 deprive an IgG concentrate of beneficial antibodies relevant for some infectious diseases. 475 When choosing affinity resins for immunoadsorption, there might be another aspect of impact on 476 IgG concentrate worth to consider. Although they are NAbs, alloanti-A and –B antibodies can be 477 induced by feeding bacteria bearing the corresponding carbohydrate epitopes [119] and are 478 considered to participate in primary host-defence. A complete removal by immunoaffinity might 479 influence some effector function of an IgG concentrate. Other antibodies possibly involved in 480 primary host defense are the anti-Gal NAbs. They show a broad specificity and can react with a 481 number of related αGal-terminated oligosaccharides, including those on bacteria [127]. Again, 482 immunoadsorption might diminish the potential of an IgG concentrate to mediate primary host 483 defense. 484 485

12

In summary, the principles of avoiding co-fractionation through cold-ethanol fractionation [128] 486 versus immune-affinity removal of histo-blood group alloantibodies can have an impact on the 487 presence of homeostatic and first-line defense antibodies. According to present knowledge, only 488 resins coated with the corresponding tetra-saccharides can ascertain the selective removal of histo-489 blood group alloantibodies presumably involved in HA. Resins coated with the corresponding di- 490 and tri-saccharides might also remove blood group alloantibodies, however not selectively. Such 491 resins in addition might remove a broad range of NAbs present in IgG concentrates at relative high 492 amounts. In the literature, the use of tri-saccharide coated resin was reported [129-131]. We have 493 found no information available in the public domain indicating which type of resin is/will be used 494 for reduction of the histo-blood group alloantibodies in large scale fractionation of IgG. 495 Furthermore, we suggest that the effect of reduction of anti-A and ant-B reacting antibodies by 496 immune-affinity on the antibody repertoire of IgG concentrates can only be assessed by e.g. using 497 pathogens/commensals, which share the saccharide epitopes, that have been used to coat the affinity 498 resins or alternatively by exposing senescent RBCs stripped off the IgGs coated in vivo. Finally, 499 techniques are required, which allow detection of low affinity, high avidity NAbs. 500 501 502 5. Falling deep into a dark lunar crater – Thrombosis 503 504 IVIG-administration related AEs, including thrombosis, have been extensively described [132]. 505 Thrombotic AEs are severe AEs and patients with risk factors require a special care. Reported 506 average incidence of IVIG induced thrombosis ranges from 3 to 13% [133]. Recognized risk factors 507 for IVIG-induced thrombosis include male gender; age > 60; diabetes; renal insufficiency, 508 dyslipidemia; hypertension; immobility; coronary disease; pre-existing vascular disease, family 509 history of early thromboembolic disease; atrial fibrillation, high-dose and high-speed IVIG 510 infusions. IVIG-induced thrombosis is reported both as venous events such as thrombosis stroke, 511 pulmonary embolism (PE), deep venous thrombosis (DVT) and arterial ischemia events such as 512 myocardial infarct and stroke. The mechanisms leading to IVIG-associated thrombosis are still not 513 completely clear; three main mechanisms have been proposed, emphasizing the role of an increased 514 blood viscosity causing a hypercoagulable state [134], the role of anticardiolipin antibodies 515 passively transferred through IVIG [135] and the role of factor XIa or other biologically highly 516 active factors passively transferred via IgG concentrates, such as PKA. Avoiding activated 517 coagulation factors in IgG concentrates starts with appropriate anticoagulation of donated 518 blood/plasma, i.e. careful mixing of anticoagulant and sample over the whole donation process. 519 Alterations in an established manufacturing process neglecting appropriate controls can also lead to 520 increased risk of transmission of activated coagulation factors. High molecular weight proteins 521 passively transferred by IVIG are probably responsible for this phenomenon [136]. In patients with 522 other risk factors, such as vascular disease, this increase in blood viscosity can precipitate 523 thromboembolic events. As elderly individuals are prone for such AEs, we like to point to the 524 possibility of elevated altered/senescent self-reacting with infused homeostatic NAbs being a 525 possible factor facilitating thrombotic events. A relationship between IVIG administration and 526 cerebral vasospasm has also been suggested by Sztajzel et al [137]; blood viscosity is a determinant 527 for oxygen delivery to the tissues, and changes in viscosity can lead to a reduction in cerebral or 528 myocardial perfusion. 529 530 We systematically reviewed case reports related to IVIG-induced thrombosis from 1986 to 2014 531 (Fig. 8). Literature search identified 35 articles containing reports concerning 65 patients [133,138-532 168]. When data were available, diagnosis, risk factors, the number of IVIG infusion prior to 533 thrombosis event and outcome have been indicated. Baseline characteristics of the patients are 534 shown in Table 3. High-dose IVIG induced thromboembolic events in 59 patients low to medium 535 IVIG doses. Mary et al. observed that the frequency and type of arterial events was inversely related 536

13

to the time elapsed from IVIG infusion; almost 50% (23 vs 21 reports) of arterial ischemic events 537 occurred in 12 hours following IVIG, while about 75% of venous thrombosis occurred after more 538 than 24 hours. No correlation between number of infusions and occurrence of adverse event was 539 observed. The main risk factors observed in this review were hypertension (19 cases, 33% of 540 prevalence), previous vascular disease (18%), and dyslipidemia (17%). Average mortality for 541 thrombotic events was 10% (arterial ischemia 9% vs venous thrombosis 11%, pulmonary embolism 542 representing the main venous fatal event). Predicting IVIG induced thrombosis is difficult. Risk 543 factors should be assessed for each patient including instrumental exams when needed. Doppler 544 ultrasound can be useful as early diagnostic tool for thrombosis or to detect the presence of 545 abnormal blood flow especially after prolonged immobility. IVIG should be administered at low 546 infusion rate to reduce the risk. The administration of antiplatelet or anticoagulant prophylaxis was 547 suggested in patients with several risk factors [147]. However, thrombotic events have been 548 reported even after several previous uncomplicated courses of treatment. In such cases, patients 549 should be examined for signs and symptoms of thrombosis during each courses of IVIG. 550 551 IgG concentrates are widely acknowledged to offer a safe, high-dose, long-term therapy option for a 552 variety of diseases. Adverse events occur rarely and mainly are mild to moderate. Deviations from 553 this rule of thumb are addressed by authorities and the plasma fractionation industry to achieve 554 corrections. Above we have reviewed two types of AE which have shown elevated frequency in the 555 near past. We tried to give some insights which might help in reducing frequencies of AEs bed side. 556 557 558 Acknowledgments 559 560 Authors are deeply grateful to Hans-Hartmut Peter, Freiburg, Germany, for his careful reading of 561 the manuscript, the valuable comments and the correction of English. 562

563

14

Legends to the figures 564 565 Figure 1: Provoking the dark. Administration of an IgG concentrate inevitably results in the 566 interactions of the exogenous IgG with the various parts of the immune system of the recipient and 567 vice versa. The rate of increase over time of exogenous IgG in the circulation of the recipient is a 568 key parameter for intensity of these interactions and eventual AEs. IgG in the circulation over 569 time (insert) is dependent on infusion rate together with the strength of the solution applied and 570 the mode of application, i.e. intravenous (solid and dashed line) or subcutaneous (dashed-dotted 571 line). From the patient’s side the threshold for a clinically noticeable AE (dotted line) might depend 572 on factors such as levels of primed cells, the efficiency of control mechanism and the metabolism of 573 e.g. mediators of inflammation. Although the area under the curve of generated e.g. complement 574 activation products and/or pro-inflammatory mediators might be the same for low (dashed line) and 575 high (solid line) infusion rates, at high infusion rate, the system might be not be able to cope with 576 the acute onset of reactions. At low infusion rates (dashed line) all the events might remain below 577 the threshold of clinically observable AE. Indeed, in several dozen of infusions using a marketed 578 IVIG in all cases except one, a more or less pronounced increase of TNF was measured at 2.5 579 hours post initiation of infusion. The only person in the cohort not showing a measurable TNF 580 increase was a woman caring at home for her brother to with full blown AIDS [170]. 581 Subcutaneously applied IgG concentrate reaches the circulation slowly (dashed-dotted line) and 582 systemic AEs are less frequent than with IVIG but they are not absent [171-177] (the case of 583 inappropriate use of SCIG is not considered). In contrast local mild to moderate AEs are more 584 frequent with SCIG [178]. 585 586 Figure 2: Staying within a network is helpful for staying at the sunny side of the moon - from 587 blood/plasma collection to blood/plasma products. Handling of blood/plasma products to obtain 588 therapeutic goods has to be performed within a regulatory framework. Manufacturing of stable 589 blood product has strictly to adhere to current Good Manufacturing Practice (cGMP). Not following 590 cGMP can lead to withdraw of a plasma product from the market from one to the other day. At 591 collection of blood/plasma screening for high titre anti-A has been introduced for one IgG 592 concentrate [112]. 593 594 Figure 3: Active measures to stay on the sunny side of the moon - possible sites for introducing 595 validated pathogen reduction and/or inactivation measures (*) along the various fractionation 596 methods. One manufacturer has already introduced an immunoaffinity chromatographic step to 597 reduce anti-A and anti-B titres in its product and another manufacturer has announced the 598 introduction of such a step to its manufacturing process [110-113] 599 600 Figure 4: An individual’s slithering into the dark. A female patient has been suffering from 601 recurrent airway infections since adolescence, occasionally complicated by pneumonia. At age 34, 602 selective IgA deficiency was diagnosed. Ten years later, she was hospitalized with pneumonia. 603 Within these 10 years her serum IgG had dropped from 7 g/L to 0.87 g/L (dashed line) and IgA was 604 undetectable. The diagnosis was changed into CVID, and IVIG replacement therapy was initiated. 605 Events on the day of infusion between 8:38 a.m. and 8:30 p.m. were as follows: two minutes after 606 the start of the infusion, having received 8 drops of a 3% IgG solution (IgA <1.2 g/L; 3% solution), 607 she experienced a flush, back pain, rigors, difficulty in breathing, and hypotension. The infusion 608 was immediately stopped. After approximately one hour the reaction has weaned, and two hours 609 later the patient felt well again, and the infusion of total 6 g IgG could be continued without further 610 complications. The patient repeatedly assured having never received any blood or plasma products 611 in the past. Notwithstanding, a serum sample drawn before the initiation of the infusion 612 demonstrated the presence of anti-IgA (full line). The follow-up of anti-IgA titers confirmed a true 613 anaphylactoid reaction mediated by anti-IgA, as the anti-IgA became undetectable immediately 614

15

after the infusion and showed a rebound phenomenon after one month. True anaphylactoid reaction 615 was also confirmed by follow-up of total complement hemolytic activity (CH50) on the day of 616 infusion (dashed-dotted line). Interestingly enough the CH50 value reached its nadir at the end of 617 the infusion when the patient had no complains. 618 619 Figure 5: Clinical signs of the ‘phlogiston’ of the dark. A CVID patient received his firs infusion of 620 IVIG (a 3% solution). Despite the start of the infusion at an infusion rate (IR) of only 10 drops per 621 min which was incrementally increased by 5 drops all 30 min, rise of heart rate (HR, Panel A), and 622 of body temperature (BT, Panel B) after 2 hours of the initiation of infusion indicated the onset of a 623 ‘phlogistic’ reaction and infusion was stopped. Stop of the infusion for 30 min immediately let drop 624 HR and BT. Restarting the infusion showed some negative effect. The first day a total of 9 g IgG 625 was infused. The next day the rapid infusion of additional 9g of IgG was without consequence on 626 BT and HR, indicating silencing of cells releasing mediators of inflammation. 627 628 Figure 6. The missing oxygen on the dark side: Ig-induced hemolysis in patients on Ig treatment. 629 Black bars indicate the number of patients with a given clinical condition 630 631 Figure 7. Monitoring for evidence for oxygen missing at the dark side. Diagnostic algorithm for Ig-632 induced hemolysis according to criteria elaborated by the Canadian Group [66]. 633 634 Figure 8. Falling deep into a dark lunar crater while being on Ig treatment. Black bars indicate the 635 number of patients with a given clinical condition and Ig-mediated thrombosis. 636

637

16

References 638 639 1. John TJ, Ninan GT, John F, Flewett TH, Francis DP, Zuckerman DP. Epidemic hepatitis B 640 caused by commercial human immunoglobulin. Lancet (1979) I:1074. 641 642 2. Barandun S, Kistler P, Jeunet F, Isliker H. Intravenous administration of human γ-globulin. Vox 643 Sang (1962) 7:157-74. 644 645 3. Ellis EF, Henney CS. Adverse reactions following administration of human gamma-globulin. 646 J.Allergy (1969) 43:45-54. 647 648 4. Kamme C, Dahlquist E, Jonsson S, Lindström F. IgG antibodies to IgA in two patients with 649 hypogammaglobulinemia treated with commercial gammaglobulin. Acta Pathol Microbiol Immunol 650 Scand (1975) 83:189-94. 651 652 5. Alving BM, Tankersley DL, Mason BL, Rossi F, Aronson DL, Finlayson JS. Contact-activated 653 factors: contaminants of immunoglobulin preparations with coagulant and vasoactive properties. J 654 Lab Clin Med (1980) 96:334-46. 655 656 6. Woodruff RK, Grigg AP, Firkin FC, Smith IL. Fatal thrombotic events during treatment of 657 autoimmune thrombocytopenia with intravenous immunoglobulin in elderly patients. Lancet (1986) 658 II:217-8. 659 660 7. Reinhart WH, Berchtold PE. Effect of high-dose intravenous immunoglobulin therapy on blood 661 rheology. Lancet (1992) 339:662-4. 662 663 8. Späth PJ, Van Holten RW, Kempf C. Pathogen safety of immunoglobulin preparations. In: Wahn 664 V, Orange J, editors. Clinical Use of Immunoglobulins. 2nd ed. Bremen - London - Boston: UNI-665 MED Verlag; (2013) pp. 26-50. 666 667 9. Azhar EI, El-Kafrawy SA, Farraj SA, Hassan AM, Al-Saeed MS, Hashem AM, et al. Evidence 668 for camel-to-human transmission of MERS coronavirus. N Engl J Med (2014) 371:1360. doi: 669 10.1056/NEJMoa1401505 670 671 10. Jackson GS, Burk-Rafel J, Edgeworth JA, Sicilia A, Abdilahi S, Korteweg J, et al. Population 672 screening for variant Creutzfeldt-Jakob disease using a novel blood test: diagnostic accuracy and 673 feasibility study. JAMA Neurol (2014) 71:421-8. doi: 10.1001/jamaneurol.2013.6001 674 675 11. Altznauer F, von Gunten S, Späth P, Simon HU. Concurrent presence of agonistic and 676 antagonistic anti-CD95 autoantibodies in intravenous Ig preparations. J Allergy Clin Immunol 677 (2003) 112:1185-90. 678 679 12. Negi VS, Elluru S, Sibéril S, Graff-Dubois S, Mouthon L, Kazatchkine MD, et al. Intravenous 680 immunoglobulin: An update on the clinical use and mechanisms of action. J Clin Immunol (2007) 681 27:233-45. 682 683 13. Barandun S, Morell A. Adverse reactions to immunoglobulin preparations. In: Nydegger, U. E. 684 editor. Immunthemotherapy. A guide to immunglobulin prophylaxis and therapy. Academic Press, 685 London, (1981) pp 223-7. 686 687

17

14. Björkander J, Wadsworth C, Hanson LA. 1040 prophylactic infusions with an unmodified 688 intravenous immunoglobullin product causing few side-effects in patients with antibody deficiency 689 syndromes. Infect (1985) 13:102-10. 690 691 15. Rachid R, Bonilla FA. The role of anti-IgA antibodies in causing adverse reactions to gamma 692 globulin infusion in immunodeficient patients: A comprehensive review of the literature. J Allergy 693 Clin Immunol (2012) 129:628-34. doi: 10.1016/j.jaci.2011.06.047 694 695 16. Lu P, Ling B, Li R. Prevalence of immunoglobulin a deficiency (IgAD) in Shanghai blood 696 donors and efforts to establish a rare blood bank of IgAD in Shanghai. Transfusion (2013) 53:98A-697 9A 698 699 17. IUIS Scientific Committee. Primary immunodeficiency diseases. Report of an IUIS Scientific 700 Committee. International Union of Immunological Societies. Clin Exp Immunol (1999) 118(Suppl 701 1):1-28. 702 703 18. Hammarström L, Persson MAA, Smith CIE. Anti-IgA in selective IgA deficiency - In vitro 704 effects and Ig subclass pattern of human anti-IgA. Scand J Immunol (1983) 18:509-13. 705 706 19. Björkander J, Hammarström L, Smith CI, Buckley RH, Cunningham-Rundles C, Hanson LA. 707 Immunoglobulin prophylaxis in patients with antibody deficiency syndromes and anti-IgA 708 antibodies. J Clin Immunol (1987) 7:8-15. 709 710 20. Ferreira A, Garcia Rodriguez MC, Fontán G. Follow-up of anti-IgA antibodies in primary 711 immunodeficient patients treated with gamma-globulin. Vox Sang (1989) 56:218-22. 712 713 21. Burks AW, Sampson HA, Buckley RH. Anaphylactic reactions after gamma globulin 714 administration in patients with hypogammaglobulinemia: Detection of IgE antibodies to IgA. N 715 Engl J Med (1986) 314:560-4. 716 717 22. Tinegate HN, Ball J, Poles D, Regan F, Sewell C, Bolton-Maggs P. Management of 718 immunoglobulin A deficiency: Lessons from haemovigilance. (2013) Vox Sang 105(S1):23 719 720 23. Palmer DS, O'Toole J, Montreuil T, Scalia V, Yi QL, Goldman M, et al. Screening of Canadian 721 Blood Services donors for severe immunoglobulin A deficiency. Transfusion (2010) 50:1524-31. 722 doi: 10.1111/j.1537-2995.2010.02588.x 723 724 24. Ropars C, Müller A, Paint N, Beige D, Avenard G. Large scale detection of IgA deficient blood 725 donors. J Immunol Meth (1982) 54:183-9. 726 727 25. Strobel E, von Meyer A. Unexpected reactions of the anti-IgA antibody particle gel 728 immunoassay. Transfus Med (2014) 24:55-7. doi: 10.1111/tme.12094 729 730 26. Robitaille N, Delage G, Long A, Thibault L, Robillard P. Allergic transfusion reactions from 731 blood components donated by IgA-deficient donors with and without anti-IgA: a comparative 732 retrospective study. Vox Sang (2010) 99:136-41. doi: 10.1111/j.1423-0410.2010.01326.x 733 734 27. Horn J, Thon V, Bartonkova D, Salzer U, Warnatz K, Schlesier M, et al. Anti-IgA antibodies in 735 common variable immunodeficiency (CVID): diagnostic workup and therapeutic strategy. Clin 736 Immunol (2007) 122:156-62. DOI: 10.1016/j.clim.2006.10.002 737 738

18

28. Sundin U, Nava S, Hammarström L. Induction of unresponsiveness against IgA in IgA-deficient 739 patients on subcutaneous immunoglobulin infusion therapy. Clin Exp Immunol (1998) 112:341-6. 740 741 29. van der Heijden J, Geissler J, van Mirre E, van Deuren M, van der Meer JWM, Salama A, et al. 742 A novel splice variant of FcgRIIa: A risk factor for anaphylaxis in patients with 743 hypogammaglobulinemia. J Allergy Clin Immunol (2013) 131:1408-16 - e5. doi: 744 10.1016/j.jaci.2013.02.009 745 746 30. Bolli R, Brügger R, Hodler G, Maeder W, Spycher MO, Gennari K. IgG dimers in liquid 747 intravenous immunoglobulin preparations. In: Kazatchkine MD, Morell A, editors. Intravenous 748 Immunoglobulin - Research and Therapy. The Parthenon Publishing Group, London, (1996) pp 749 307-8. 750 751 31. Schnorf J, Arnet B, Burek-Kozlowska A, Gennari K, Rohner R, Späth PJ et al. Laboratory 752 parameters measured during infusion of immunoglobulin preparations for intravenous use and 753 related tolerability. In: Kazatchkine MD, Morell A, editors. Intravenous Immunoglobulin - 754 Research and Therapy. The Parthenon Publishing Group, London, (1996) pp 312-3. 755 756 32. Spycher, M. O., Bolli, R., Hodler, G., Gennari, K., Hubsch, A, Späth, P., and Schnorf, J. Well-757 tolerated liquid intravenous immunoglobulin G preparations (IVGG) have a low immunoglobulin G 758 dimer (IgG-dimer) content. Journal of Autoimmunity (1999) 96 (Suppl 1):S96A. 759 760 33. Bleeker WK, Agterberg J, Rigter G, de Vries van Rosen A, Bakker JC. An animal model for the 761 detection of hypotensive side effects of immunoglobulin preparations. Vox Sang (1987) 52:281-90. 762 763 34. Bleeker WK, Agterberg J, Rigter G, Van Rooijen N, Bakker JC. Key role of macrophages in 764 hypotensive side effects of immunoglobulin preparations. Studies in an animal model. Clin Exp 765 Immunol (1989) 77:338-44. 766 767 35. Bleeker WK, Teeling JL, Verhoeven AJ, Rigter GM, Agterberg J, Tool AT, et al. Vasoactive 768 side effects of intravenous immunoglobulin preparations in a rat model and their treatment with 769 recombinant platelet-activating factor acetylhydrolase. Blood (2000) 95:1856-61. 770 771 36. Teeling JL, Bleeker WK, Rigter GM, Van Rooijen N, Kuijpers TW, Hack CE. Intravenous 772 immunoglobulin preparations induce mild activation of neutrophils in vivo via triggering of 773 macrophages - studies in a rat model. Br J Haematol (2001) 112:1031-40. 774 775 37. Kroez M, Kanzy EJ, Gronski P, Dickneite G. Hypotension with intravenous immunoglobulin 776 therapy: importance of pH and dimer formation. Biologicals (2003) 31:277-86. 777 778 38. Teeling JL, Jansen-Hendriks T, Kuijpers TW, De Haas M, van de Winkel JG, Hack CE, et al. 779 Therapeutic efficacy of intravenous immunoglobulin preparations depends on the immunoglobulin 780 G dimers: studies in experimental immune thrombocytopenia. Blood (2001) 98:1095-9. 781 782 39. Teeling JL, Bleeker WK, Rigter GM, Van Rooijen N, Kuijpers TW, Hack CE. Intravenous 783 immunoglobulin preparations induce mild activation of neutrophils in vivo via triggering of 784 macrophages - studies in a rat model. Br J Haematol (2001) 112:1031-40. 785 786 40. Farber CM, Crusiaux A, Schandene L, Van Vooren JP, Goldman M, Dupont E, et al. Tumor 787 necrosis factor and intravenous gammaglobulins in common variable immunodeficiency. Clin 788 Immunol Immunopathol (1994) 72:233-6. 789

19

790 41. Bagdasarian A, Tonetta S, Harel W, Mamidi R, Uemura Y. IVIG adverse reactions: potential 791 role of cytokines and vasoactive substances. Vox Sang (1998) 74:74-82. 792 793 42. Michelis FV, Branch DR, Scovell I, Bloch E, Pendergrast J, Lipton JH, et al. Acute hemolysis 794 after intravenous immunoglobulin amid host factors of ABO-mismatched bone marrow 795 transplantation, inflammation, and activated mononuclear phagocytes. Transfusion (2014) 54:681-796 90. 797 798 43. Ling ZD, Yeoh E, Webb BT, Farrell K, Doucette J, Matheson DS. Intravenous immunoglobulin 799 induces interferon- and interleukin-6 in vivo. J Clin Immunol (1993) 13:302-9. 800 801 44. Yoo EM, Wims LA, Chan LA, Morrison SL. Human IgG2 can form covalent dimers. J 802 Immunol (2003) 170:3134-8. 803 804 45. Vassilev TL, Bineva IL, Dietrich G, Kaveri SV, Kazatchkine MD. Variable region-connected, 805 dimeric fraction of intravenous immunoglobulin enriched in natural autoantibodies. J Autoimmun 806 (1995) 8:405-13. 807 808 46. Simon HU, Späth PJ. IVIG - Mechanisms of action. Allergy (2003) 58:543-52. 809 810 47. Gronski P. IgG dimers in multidonor-derived immunoglobulins: Aspects of generation and 811 function. Curr Pharm Des (2006) 12:181-90. 812 813 48. Tankersley DL, Preston MS, Finlayson JS. Immunoglobulin G dimer: An idiotype-anti-idiotype 814 complex. Mol Immunol (1988) 25:41-8. 815 816 49. Roux KH, Tankersley DL. A view of the human idiotypic repertoire - Electron microscopic and 817 immunologic analyses of spontaneous idiotype-anti-idiotype dimers in pooled human IgG. J 818 Immunol (1990) 144:1387-95. 819 820 50. Gronski P, Schridde C, Forsterling HD. Polyreactive antibodies in multidonor-derived 821 immunoglobulin G: theory and conclusions drawn from experiments. Immunobiology (2010) 822 215:356-69. doi: 10.1016/j.imbio.2009.06.015. 823 824 51. Miescher SM, Schaub A, Ghielmetti M, Baumann M, Vogel M, Bolli R, et al. Comparative 825 analysis of antigen specificities in the monomeric and dimeric fractions of intravenous 826 immunoglobulin. Ann NY Acad Sci (2005) 1051:582-90. 827 828 52. Wymann S, Ghielmetti M, Schaub A, Baumann MJ, Stadler BM, Bolli R, et al. Monomerization 829 of dimeric IgG of intravenous immunoglobulin (IVIg) increases the antibody reactivity against 830 intracellular antigens. Mol Immunol (2008) 45:2621-8. doi: 10.1016/j.molimm.2007.12.020. 831 832 53. Relkin NR, Szabo P, Rotondi M, Mujalli D. Antibodies in the dimer fraction of IVIg have the 833 capacity to bind beta amyloid. Alzheimer's Dementia (2009) 5:427-8. 834 835 54. Schaub A, von Gunten S, Vogel M, Wymann S, Stadler B, Spycher M, et al. An analysis of 836 anti-Fas and anti-Siglec-9 autoantibodies in monomeric and dimeric fractions of IVIG. Allergy 837 (2009) 64:268-9. 838 839

20

55. Schaub A, von Gunten S, Vogel M, Wymann S, Rüegsegger M, Stadler BM, et al. Dimeric 840 IVIG contains natural anti-Siglec-9 autoantibodies and their anti-idiotypes. Allergy (2011) 66:1030-841 7. doi: 10.1111/j.1398-9995.2011.02579.x. 842 843 56. Bridgham, M., Drake, M., and Maguire, K. A case of haemolysis following administration of 844 intravenous immunoglobulin. Transfus Med (2014) 24:58. 845 846 57. Clemenz MR, Joseph WM, Shuler MJ, Lynn AW. Intravenous immunoglobulin-induced 847 hemolytic anemia in a patient with juvenile dermatomyositis. J Drugs Dermatol (2013) 12:111-3. 848 849 58. Desborough MJ, Miller J, Thorpe SJ, Murphy M F, Misbah SA. et al. Intravenous 850 immunoglobulin-induced haemolysis: a case report and review of the literature. Transfus Med 851 (2013) 24:219-26. doi: 10.1111/tme.12083 852 853 59. Mohamed M, Bates G, Eastley B. Massive intravascular haemolysis after high dose intravenous 854 immunoglobulin therapy. Br J Haematol (2013) 160:570. doi: 10.1111/bjh.12182. 855 856 60. Rink BD, Gonik B, Chmait RH, O'Shaughnessy R. Maternal hemolysis after intravenous 857 immunoglobulin treatment in fetal and neonatal alloimmune thrombocytopenia. Obstet Gynecol 858 (2013) 121:471-3. doi: 10.1097/AOG.0b013e3182765c63. 859 860 61. Berard R, Whittemore B, Scuccimarri R. Hemolytic anemia following intravenous 861 immunoglobulin therapy in patients treated for Kawasaki disease: a report of 4 cases. Pediatr 862 Rheumatol Online J (2012) 10:10. doi: 10.1186/1546-0096-10-10. 863 864 62. Michelis FV, Branch DR, Scovell I, Bloch E, Pendergrast J, Lipton JH, et al. Acute hemolysis 865 after intravenous immunoglobulin amid host factors of ABO-mismatched bone marrow 866 transplantation, inflammation, and activated mononuclear phagocytes. Transfusion (2014) 54:681-867 90. doi: 10.1111/trf.12329. 868 869 63. Pintova S, Bhardwaj AS, Aledort LM. IVIG--a hemolytic culprit. N Engl J Med (2012) 870 367:974-6. doi: 10.1056/NEJMc1205644. 871 872 64. Morgan S, Sorensen P, Vercellotti G, Zantek ND. Haemolysis after treatment with intravenous 873 immunoglobulin due to anti-A. Transfus Med (2011) 21:267-70. doi: 10.1111/j.1365-874 3148.2011.01078.x. 875 876 65. Welles CC, Tambra S, Lafayette RA. Hemoglobinuria and acute kidney injury requiring 877 hemodialysis following intravenous immunoglobulin infusion. Am J Kidney Dis (2010) 55:148-51. 878 doi: 10.1053/j.ajkd.2009.06.013. 879 880 66. Canadian Blood Services. Important information regarding IVIG associated hemolysis. 881 Available: 882 www.bloodservices.ca/CentreApps/Internet/UW_V502_MainEngine.nsf/resources/CustomerLetters883 09/$file/CL_(2009)-02.pdf (accessed October 2014). 884 885 67. Gordon DJ, Sloan SR, de Jong JL. A pediatric case series of acute hemolysis after 886 administration of intravenous immunoglobulin. Am J Hematol (2009) 84:771-2. doi: 887 10.1002/ajh.21544. 888 889

21

68. Kahwaji J, Barker E, Pepkowitz S, Klapper E, Villicana R, Peng A, Chang R, Jordan SC, Vo 890 AA. Acute hemolysis after high-dose intravenous immunoglobulin therapy in highly HLA 891 sensitized patients. Clin J Am Soc Nephrol (2009) 4:1993-7. doi: 10.2215/CJN.04540709. 892 893 69. Daw Z, Padmore R, Neurath D, Cober N, Tokessy M, Desjardins D, et al. Hemolytic 894 transfusion reactions after administration of intravenous immune (gamma) globulin: a case series 895 analysis. Transfusion (2008) 48:1598-1601 doi: 10.1111/j.1537-2995.2008.01721.x. 896 897 70. Yin F, Nesbitt JA, Tobian AA, Holt PA, Mikdashi J. Hemolytic anemia following intravenous 898 immunoglobulin administration. Am J Hematol (2008) 83:825. doi: 10.1002/ajh.21263. 899 900 71. Coghill J, Comeau T, Shea T, Braddy L, Bandarenko N, Afenyi-Annan A, Harvey D. Acute 901 hemolysis in a patient with cytomegalovirus pneumonitis treated with intravenous immunoglobulin 902 (IVIG). Biol Blood Marrow Transplant (2006) 12:786-8. 903 904 72. Singh-Grewal D, Kemp A, Wong MA. prospective study of the immediate and delayed adverse 905 events following intravenous immunoglobulin infusions. Arch Dis Child (2006) 91:651-4. 906 907 73. Chamouni P, Tamion F, Gueit I, Girault C, Lenain P, Varin R, Czernichow P. Adverse effect of 908 polyvalent immunoglobulin in the treatment of Guillain-Barré syndrome. Transfus Apher Sci (2003) 909 28:117-24. 910 911 74. Karaaslan S, Oran B, Kalisman U. Hemolysis After Administration of High-Dose 912 Immunoglobulin in a Patient with Myocarditis. Turk J Haematol (2003) 20:237-40. 913 914 75. Trifa M, Simon L, Hamza J, Bavoux F, des Roziers NB. Haemolytic anaemia associated with 915 high dose intravenous immunoglobulin therapy in a child with Guillain-Barré syndrome. Arch Dis 916 Child (2003) 88:836-7. 917 918 76. Ballot-Brossier C, Mortelecque R, Sinegre M, Marceau A, Dauriat G, Courtois F. Insisting on 919 intravenous polyvalent immunoglobulin therapy in polymyositis in spite of the occurrence of sever 920 hemolytic anemia – Poursuite du traitement d'une polymyosite par les immunoglobulines 921 intraveineuses polyvalentes malgre la survenue d'une anémie hémolytique sévère. Transfus Clin 922 Biol (2001) 8:94-9. 923 924 77. Nagakawa M, Watanabe N, Okuno M, Kondo M, Okagawa H, Taga T. Severe hemolytic 925 anemia following high-dose intravenous immunoglobulin administration in a patient with Kawasaki 926 disease. Am J Hematol (2000) 63:160-1. doi: 10.1002/(SICI)1096-8652(200003)63:3<160. 927 928 78. Wilson JR, Bhoopalam H, Fisher M. Hemolytic anemia associated with intravenous 929 immunoglobulin. Muscle Nerve (1997) 20:1142-5. 930 931 79. Munoz J, Garcia A, Jordan C, Rubio A, Ferran C, Martinez Matos JA. Hemolysis during 932 immunoglobulin therapy - Hemolisis durante el tratamiento con inmunoglobulinas. Sangre (Barc.) 933 (1996) 41:72-3. 934 935 80. Tamada K, Kohga M, Masuda H, Hattori K. Hemolytic anemia following high-dose intravenous 936 immunoglobulin administration. Acta Paediatr Jpn (1995) 37:391-3. 937 938

22

81. Boothe G, Brecher ME, Root M, Robinson J, Haley R. Acute hemolysis due to passively 939 transfused high-titer anti-B causing spontaneous in vitro agglutination. Immunohematology (1995) 940 11:43-5. 941 942 82. Thomas MJ, Misbah SA, Chapel HM, Jones M, Elrington G, Newsom-Davis J. Hemolysis after 943 high-dose intravenous Ig. Blood (1993) 82:3789. 944 945 83. Comenzo RL, Malachowski ME, Meissner HC, Fulton DR, Berkman EM. Immune hemolysis, 946 disseminated intravascular coagulation, and serum sickness after large doses of IVIG for Kawasaki 947 disease. J Pediatr (1992) 120:926-8. 948 949 84. Rovelli A, D' Angelo P, Balduzzi A, Borzini P, Biondi A, Uderzo C. Acute intravascular 950 haemolysis associated with high dose immunoglobulin after bone marrow transplantation for acute 951 myelogenous leukemia. Leuk Lymphoma (1991) 5:71-4. 952 953 85. Okubo S, Ishida T, Yasunaga K. Hemolysis after intravenous immune globulin therapy: relation 954 to IgG subclasses of red cell antibody. Transfusion (1990) 30:436-8. 955 956 86. Hillyer CD, Schwenn MR, Fulton DR, Meissner HC, Berkman EM. et al. Autoimmune 957 hemolytic anemia in Kawasaki disease: a case report. Transfusion (1990) 30:738-40. 958 959 87. Nicholls MD, Cummins JC, Davies VJ, Greenwood JK. Haemolysis induced by intravenously-960 administered immunoglobulin. Med J Aust (1989) 150:404-6. 961 962 88. Kim HC, Park CL, Cowan JH 3rd, Fattori FD, August CS. Massive intravascular hemolysis 963 associated with intravenous immunoglobulin in bone marrow transplant recipients. Am J Pediatr 964 Hematol Oncol (1988) 10:69-74. 965 966 89. Brox AG, Cournoyer D, Sternbach M, Spurll G. Hemolytic anemia following intravenous 967 gamma globulin administration. Am J Med (1987) 82:633-5. 968 969 90. Copelan EA, Strohm PL, Kennedy MS, Tutschka PJ Hemolysis following intravenous immune 970 globulin therapy. Transfusion (1986) 26:410-412. 971 972 91. Atrah HI, Crawford RJ, Templeton JG, Carlyle JE. Transient haemoglobin drop following high 973 dose intravenous immunoglobulin. Clin Lab Hematol (1985) 7:283. 974 975 92. Kessary-Shoham H, Levy Y, Shoenfeld Y, Lorber M, Gershon H. In vivo administration of 976 intravenous immunoglobulin (IVIg) can lead to enhanced erythrocyte sequestration. J Autoimmun 977 (1999) 13:129-35. 978 979 93. Quinti I, Pulvirenti F, Milito C, Granata G, La Marra F, Farrugia A, Coluzzi S, Girelli G. 980 Hemolysis in patients with antibody deficiencies on immunoglobulin replacement treatment. 981 Transfusion (2014) in press. 982 983 94. Thorpe SJ, Fox BJ, Dolman CD, Lawrence J, Thorpe R. Batches of intravenous 984 immunoglobulin associated with adverse reactions in recipients contain atypically high anti-Rh D 985 activity. Vox Sang (2003) 85:80-4. 986 987 95. Baxley A, Akhtari M. Hematologic toxicities associated with intravenous immunoglobulin 988 therapy. Internatl Immunopharmacol (2011) 11:1663–7. doi: 10.1016/j.intimp.2011.07.024. 989

23

990 96. Kahwaji J, Barker E, Pepkowitz S, Klapper E, Villicana R, Peng A, Chang R, Jordan SC, Vo 991 AA. Acute hemolysis after high-dose intravenous immunoglobulin therapy in highly HLA 992 sensitized patients. Clin J Am Soc Nephrol (2009) 4:1993-7. doi: 10.2215/CJN.04540709. 993 994 97. Thorpe SJ, Fox BJ, Dolman CD, Thorpe R. Anti-A and anti-B activity in batches of different 995 intravenous immunoglobulin products determined using a direct haemagglutination method. 996 Biologicals (2005) 33:111-6. 997 998 98. Thorpe SJ, Fox B, Sharp G, Virata ML, Yu MW, Thorpe R. International collaborative study to 999 evaluate candidate reference reagents to standardize haemagglutination testing for anti-A and anti-B 1000 in normal intravenous immunoglobulin products. Vox Sang (2009) 97:160-8. 1001 1002 99. European Directorate for the Quality of Medicines and Healthcare. Human normal 1003 immunoglobulin for intravenous administration. In: European Pharmacopoeia (7th Monograph 1004 0918 edn), (2011), 4166–8. 1005 1006 100. Council of Europe, Strasbourg, France; European Directorate for the Quality of Medicines and 1007 Healthcare. Anti-A and anti-B haemagglutinins. In: European Pharmacopoeia (7th General chapter 1008 2.6.20 edn). (2011). 1009 1010 101. Council of Europe, Strasbourg, France; European Directorate for the Quality of Medicines and 1011 Healthcare. Test for anti-D antibodies in human immunoglobulin. In: European Pharmacopoeia 1012 (7th General chapter 2.6.26 edn). (2011), 3546. Council of Europe, Strasbourg, France. 1013 1014 102. Shoham-Kessary H, Naot Y, Gershon H. Immune complex-like moieties in immunoglobulin 1015 for intravenous use (i.v.Ig) bind complement and enhance phagocytosis of human erythrocytes. Clin 1016 Exp Immunol (1998) 113:77-84. 1017 1018 103. Kessary-Shoham H, Levy Y, Shoenfeld Y, Lorber M, Gershon,H. In vivo administration of 1019 intravenous immunoglobulin (IVIg) can lead to enhanced erythrocyte sequestration. J Autoimmun 1020 (1999) 13:129-35. 1021 1022 104. Hudson KE, Lin E, Hendrickson JE, Lukacher AE, Zimring JC. Regulation of primary 1023 alloantibody response through antecedent exposure to a microbial T-cell epitope. Blood (2010) 1024 115:3989-3996. doi: 10.1182/blood-2009-08-238568. 1025 1026 105. Rodeghiero F, Schiavotto C, Castaman G, Vespignani M, Ruggeri M, Dini E. A follow-up 1027 study of 49 adult patients with idiopathic thrombocytopenic purpura treated with high-dose 1028 immunoglobulins and anti-D immunoglobulins. Haematologica (1992) 77:248-52. 1029 1030 106. Padmore RF. Hemolysis upon intravenous immunoglobulin transfusion. Transfus Apher Sci 1031 (2012) 46:93-6. doi: 10.1016/j.transci.2011.11.004. 1032 1033 107. Report of the FDA meeting on Strategies to address Hemolytic complications of Immune 1034 Globulin Infusions, Washington DC, (2014). 1035 1036 108. Sewell WA, Kerr J, Behr-Gross ME, Peter HH; Kreuth Ig Working Group. European 1037 consensus proposal for immunoglobulin therapies. Eur J Immunol (2014) 44:2207-14. doi: 1038 10.1002/eji.201444700 1039 1040

24

109. Jacobson, NM. The art of balanced production. In: Valverde, JL, editor. Pharmaceuticals 1041 Policy and Law - Blood, Plasma and Plasma Proteins: A Unique Contribution to Modern 1042 Healthcare. Amsterdam, NL, IOS Press. (2006) 7:81-7. 1043 1044 110. Dhainaut F, Guillaumat PO, Dib H, Perret G, Sauger A, De Coupade C, et al. In vitro and in 1045 vivo properties differ among liquid intravenous immunoglobulin preparations. Vox Sang (2013) 1046 104:115-126. doi: 10.1111/j.1423-0410.2012.01648.x 1047 1048 111. Roch B, Monrepos S, Dhainaut F, Milchaski C, De Coupade C, Sauger A, van der Meersch A, 1049 Paolantonacci P, and Rossi F. Patient safety through an IVIg mastered manufacturing process. 1050 Posters, European Symposium Proceedings (2013) 1-351. European Directorate for the Quality of 1051 Medicines & HealthCare; https://www.edqm.eu/medias/fichiers/posters_kreuth_iii.pdf (accessed 1052 October 2014). 1053 1054 112. Siani B, Willimann K, Wymann S, Marques AA, Widmer E. Isoagglutinin reduction in human 1055 immunoglobulin products by donor screening. BiolTher (2014) Epub ahead of print. 1056 1057 113. Höfferer, L. IgG product development isoagglutinin reduction measures. Oral presentation, 1058 International Plasma Protein Congress, Vienna, Austria, (2014). 1059 1060 114. Späth PJ, Lutz HU. Naturally occurring antibodies/autoantibodies in polyclonal 1061 immunoglobulin concentrates. Lutz, H. U. Naturally Occurring Antibodies (NAbs). Adv Exp Med 1062 Biol (2012) 750:239-261. doi 10.1007/978-1-4614-3461-0_18 1063 1064 115. Lutz HU BussolinobF, Flepp R, Fasler S, Stammler P, Kazatchkine M D, et al. Naturally 1065 occurring anti-band-3 antibodies and complement together mediate phagocytosis of oxidatively 1066 stressed human erythrocytes. Proc Natl Acad Sci USA (1987) 84:7368-72. 1067 1068 116. Lutz HU, Stammler P, Jelezarova E, Nater M, Späth PJ. High doses of immunoglobulin G 1069 attenuate immune aggregate-mediated complement activation by enhancing physiologic cleavage of 1070 C3b in C3bn-IgG complexes. Blood (1996) 88:184-93. 1071 1072 117. Lutz HU, Stammler P, Bianchi V, Trüeb RM, Hunziker T, Burger R. Intravenously applied 1073 IgG stimulates complement attenuation in a complement-dependent autoimmune disease at the 1074 amplifying C3 convertase level. Blood (2004) 103:465-72. 1075 1076 118. Nydegger UE, Tevaearai H, Berdat P, Rieben R, Carrel T, Mohacsi P, et al. Histo-blood group 1077 antigens as allo- and autoantigens. Ann NY Acad Sci (2005) 1050:40-51. 1078 1079 119. Springer GF, Horton RE. Blood group isoantibody stimulation in man by feeding blood group-1080 active bacteria. J Clin Invest (1969) 48:1280-91. 1081 1082 120. Spalter SH, Kaveri SV, Bonnin E, Mani JC, Cartron JP, Kazatchkine MD Normal human 1083 serum contains natural antibodies reactive with autologous AB0 blood group antigens. Blood (1999) 1084 93:4418-24. 1085 1086 121. Galili U, Rachmilewitz EA, Peleg A, Flechner I. A unique natural human IgG antibody with 1087 anti-alpha-galactosyl specificity. J Exp Med (1984) 160:1519-31. 1088 1089 122. Obukhova P, Rieben R, Bovin N. Normal human serum contains high levels of anti-Gal1-1090 4GlcNAc antibodies. Xenotransplant (2007) 14:627-35. 1091

25

1092 123. Obukhova P, Korchagina E, Henry S, Bovin N. Natural anti-A and anti-B of the AB0 system: 1093 allo- and autoantibodies have different epitope specificity. Transfusion (2012) 52:860-9. doi: 1094 10.1111/j.1537-2995.2011.03381.x 1095 1096 124. Bovin N, Bovin,N.; Obukhova,P.; Shilova,N.; Rapoport,E.; Popova,I.; Navakouski,M.; 1097 Unverzagt,C.; Vuskovic,M.; Huflejt,M. et al. Repertoire of human natural anti-glycan 1098 immunoglobulins. Do we have auto-antibodies? Biochim Biophys Acta (2012) 1820:1373-82. doi: 1099 10.1016/j.bbagen.2012.02.005 1100 1101 125. Galili U, Mandrell RE, Hamadeh RM, Shohet SB, McLeod Griffiss J. Interaction between 1102 human natural anti-alpha-galactosyl immunoglobulin G and bacteria of the human flora. Infect 1103 Immun (1988) 56:1730-7. 1104 1105 126. Sorette MP, Galili U, Clark MR. Comparison of serum anti-band 3 and anti-Gal antibody 1106 binding to density-separated human red blood cells. Blood (1991) 77:628-36. 1107 1108 127. Wieslander J, Mansson O, Kallin E, Gabrielli A, Nowack H, Timple R. Specificity of human 1109 antibodies against galα1-3gal carbohydrate epitope and distinction from natural antibodies reacting 1110 with galα1-2gal or galα1-4 gal. Glycoconj J (1990) 7:85-100. 1111 1112 128. Nardini C. Anti-A and anti-B haemagglutinin trend analysis during manufacturing process of 1113 IVIG. Presentation at Workshop “Strategies to Address Hemolytic Complications of Immune 1114 Globulin Infusions” US Food and Drug Administration. (2014). 1115 1116 129. Rieben R, Korchagina EY, von Allmen E, Hovinga JK, Lämmle,B.; Jungi TW In vitro 1117 evaluation of the efficacy and biocompatibility of new, synthetic AB0 immunoabsorbents. 1118 Transplantation (1995) 60:425-430. 1119 1120 130. Alikhani A, Korchagina EY, Chinarev AA, Bovin NV, Federspiel WJ. High molecular weight 1121 blood group A trisaccharide-polyacrylamide glycoconjugates as synthetic blood group A antigens 1122 for anti-A antibody removal devices. J Biomed Mater Res B Appl Biomater (2009) 91:845-854. 1123 1124 131. Gautam S, Korchagina EY, Bovin NV, Federspiel WJ. Specific antibody filter (SAF) binding 1125 capacity enhancement to remove anti-A antibodies. J Biomed Mater Res B Appl Biomater (2010) 1126 95:475-480. 1127 1128 132. Orbach H, Katz U, Sherer Y, Shoenfeld Y. Intravenous immunoglobulin: adverse effects and 1129 safe administration. Clin Rev Allergy Immunol (2005) 29:173-84. 1130 1131 133. Rajabally YA, Kearney DA. Thromboembolic complications of intravenous immunoglobulin 1132 therapy in patients with neuropathy: a two-year study. J Neurol Sci (2011) 308:124-7. doi: 1133 10.1016/j.jns.2011.05.035. 1134 1135 134. Dalakas MC. High-dose intravenous immunoglobulin and serum viscosity: risk of precipitating 1136 thromboembolic events. Neurology (1994) 44:223–6. 1137 1138 135. B. Sakem, K. Matozan, U.E. Nydegger, G. Weigel, A. Griesmacher, L. Risch, Anti-red blood 1139 cell antibodies, free light chains, and antiphospholipid antibodies in intravenous immunoglobulin 1140 preparations. Isr Med Assoc J (2013) 15:617-21. 1141 1142

26

136. Grosse-Wilde H, Blasczyk R, Westhoff U. Soluble HLA class I and class II concentrations in 1143 commercial immunoglobulin preparations. Tissue Antigens (1992) 39:74-7. 1144 1145 137. Sztajzel R, Le Floch-Rohr J, Eggimann P. High-dose intravenous immunoglobulin treatment 1146 and cerebral vasospasm: A possible mechanism of ischemic encephalopathy? Eur Neurol (1999) 1147 41:153-8. 1148 1149 138. Vinod KV, Kumar M, Nisar KK. High dose intravenous immunoglobulin may be complicated 1150 by myocardial infarction. Neuropediatrics (2008) 39:131-3. doi: 10.1055/s-2008-1077088. 1151 1152 139. Iroh Tam PY, Richardson M, Grewal S. Fatal case of bilateral internal jugular vein thrombosis 1153 following IVIg infusion in an adolescent girl treated for ITP. Am J Hematol (2008) 83:323-5. 1154 1155 140. Al-Riyami AZ, Lee J, Connolly M, Shereck. Cerebral sinus thrombosis following IV 1156 immunoglobulin therapy of immune thrombocytopenia purpura. Pediatr Blood Cancer (2011) 1157 57:157-9. doi: 10.1002/pbc.22968. 1158 1159 141. Sin YH, Kim YJ, Oh JS, Lee JH, Kim SM, Kim JK. Graft rupture after high-dose intravenous 1160 immunoglobulin therapy in a renal transplant patient. Nephrology (2014) 19:35-6. doi: 1161 10.1111/nep.12248. 1162 1163 142. Min J, Bhatt A, Aburashed R, Burton S. Cerebral venous and sinus thrombosis associated with 1164 subcutaneous immunoglobulin injection and oral contraceptive use. Neurol Sci (2012) 33:627-9. 1165 doi: 10.1007/s10072-011-0778-y. 1166 1167 143. Barada W, Muwakkit S, Hourani R, Bitar M, Mikati M. Cerebral sinus thrombosis in a patient 1168 with humoral immunodeficiency on intravenous immunoglobulin therapy: a case report. Indian J 1169 Crit Care Med (2014) 18:247-9. doi: 10.4103/0972-5229.130579. 1170 1171 144. Lee YJ, Shin JU, Lee J, Kim K, Kim WS, Ahn JS, Jung CW, Kang WK. A case of deep vein 1172 thrombosis and pulmonary thromboembolism after intravenous immunoglobulin therapy. J Korean 1173 Med Sci (2007) 22:758-61. 1174 1175 145. White DA, Leonard MC. Acute stroke with high-dose intravenous immune globulin. Am J 1176 Health Syst Pharm (2007) 64:1611-4. 1177 1178 146. Feuillet L, Milandre L, Ali Cherif A. Venous and arterial thrombosis following administration 1179 of intravenous immunoglobulins. Blood Coagul Fibrinolysis (2006) 17:85. 1180 1181 147. Marie I, Hervé F, Kerleau JM, Maurey G, Levesque H. Intravenous immunoglobulin-1182 associated vena cava thrombosis. Thromb Haemost (2006) 96:849-51. 1183 1184 148. Marie I, Maurey G, Hervé F, Hellot MF, Levesque H. Intravenous immunoglobulin-associated 1185 arterial and venous thrombosis; report of a series and review of the literature. Br J Dermatol (2006) 1186 155:714-21. 1187 1188 149. Hefer D, Jaloudi M. Thromboembolic events as an emerging adverse effect during high-dose 1189 intravenous immunoglobulin therapy in elderly patients: a case report and discussion of the relevant 1190 literature. Ann Hematol (2004) 83:661-5. 1191 1192

27

150. Geller JL, Hackner D. Diffuse venous thromboemboli associated with IVIg therapy in the 1193 treatment of streptococcal toxic shock syndrome: case report and review. Ann Hematol (2005) 1194 84:601-4. 1195 1196 151. Feuillet L, Guedj E, Laksiri N, Philip E, Habib G, Pelletier J, Ali Cherif. Deep vein thrombosis 1197 after intravenous immunoglobulins associated with methylprednisolone. Thromb Haemost (2004) 1198 92:662-5. 1199 1200 152. Sheehan DJ, Lesher JL. Deep venous thrombosis after high-dose intravenous immunoglobulin 1201 in the treatment of pemphigus vulgaris. Cutis (2004) 73:403-6. 1202 1203 153. Vucic S, Chong PS, Dawson KT, Cudkowicz M, Cros D Jr. Thromboembolic complications of 1204 intravenous immunoglobulin treatment. Eur Neurol (2004) 52:141-4. 1205 1206 154. Stamboulis E, Theodorou V, Kilidireas K, Apostolou T. Acute myocardial infarction following 1207 intravenous immunoglobulin therapy for chronic inflammatory demyelinating polyneuropathy in 1208 association with a monoclonal immunoglobulin G paraprotein. Eur Neurol (2004) 51:51. 1209 1210 155. Katz KA, Hivnor CM, Geist DE, Shapiro M, Ming ME, Werth VP. Stroke and deep venous 1211 thrombosis complicating intravenous immunoglobulin infusions.; Arch Dermatol (2003) 139:991-3. 1212 1213 156. Zaidan R, Al Moallem M, Wani BA, Shameena AR, Al Tahan AR, Daif AK, Al Rajeh S. 1214 Thrombosis complicating high dose intravenous immunoglobulin: report of three cases and review 1215 of the literature. Eur J Neurol (2003) 10:367-72. 1216 1217 157. Brown HC, Ballas ZK. Acute thromboembolic events associated with intravenous 1218 immunoglobulin infusion in antibody-deficient patients. J Allergy Clin Immunol (2003) 112:797-9. 1219 1220 158. Evangelou N, Littlewood T, Anslow P, Chapel H. Transverse sinus thrombosis and IVIg 1221 treatment: a case report and discussion of risk-benefit assessment for immunoglobulin treatment. J 1222 Clin Pathol (2003) 56:308-9. 1223 1224 159. Emerson GG, Herndon CN, Sreih AG. Thrombotic complications after intravenous 1225 immunoglobulin therapy in two patients. Pharmacotherapy (2002) 22:1638-41. 1226 1227 160. Alliot C, Rapin JP, Besson M, Bedjaoui F, Messouak D. Pulmonary embolism after 1228 intravenous immunoglobulin. J R Soc Med (2001) 94:187-8. 1229 1230 161. Sherer Y, Levy Y, Langevitz P, Rauova L, Fabrizzi F, Shoenfeld Y. Adverse effects of 1231 intravenous immunoglobulin therapy in 56 patients with autoimmune diseases. Pharmacology 1232 (2001) 62:133-7. 1233 1234 162. Elkayam O, Paran D, Milo R, Davidovitz Y, Almoznino-Sarafian D, Zeltser D, Yaron M, 1235 Caspi D. Acute myocardial infarction associated with high dose intravenous immunoglobulin 1236 infusion for autoimmune disorders. A study of four cases. Ann Rheum Dis (2000) 59:77-80. 1237 1238 163. Go RS, Call TG. Deep venous thrombosis of the arm after intravenous immunoglobulin 1239 infusion: case report and literature review of intravenous immunoglobulin-related thrombotic 1240 complications. Mayo Clin Proc (2000) 75:83-5. 1241 1242

28

164. Turner B, Wills AJ. Cerebral infarction complicating intravenous immunoglobulin therapy in a 1243 patient with Miller Fisher syndrome. J Neurol Neurosurg Psychiatry (2000) 68:790-1. 1244 1245 165. Harkness KA, Goulding P. Central retinal vein occlusion complicating treatment with 1246 intravenous immunoglobulin. Eye (Lond) (2000) 14:662-3. 1247 1248 166. Paolini R, Fabris F, Cella G. Acute myocardial infarction during treatment with intravenous 1249 immunoglobulin for idiopathic thrombocytopenic purpura (ITP); Am J Hematol (2000) 65:177-8. 1250 1251 167. Rosenbaum JT. Myocardial infarction as a complication of immunoglobulin therapy. Arthritis 1252 Rheum (1997) 40:1732-3. 1253 1254 168. Oh KT, Boldt HC, Danis RP. Iatrogenic central retinal vein occlusion and hyperviscosity 1255 associated with high-dose intravenous immunoglobulin administration. Am J Ophthalmol (1997) 1256 124:416-8. 1257 1258 169. Woodruff RK, Grigg AP, Firkin FC, Smith IL. Fatal thrombotic events during treatment of 1259 autoimmune thrombocytopenia with intravenous immunoglobulin in elderly patients. Lancet II 1260 (1986) 2:217-8. 1261 1262 170. Späth P, Kempf C, Gold R. Herstellung, Verträglichkeit und Virussicherheit von intravenösen 1263 Immunglobulinen. In: Berlit P, editor. Immunglobuline in der klinischen Neurologie. 1st ed.: 1264 Steinkopf, Darmstadt, Germany; (2001) pp. 1-42. 1265 1266 171. Divan HA, Menis M, Sridhar G, Selvam N, Forshee R, Izurieta HS. Occurrence of hemolytic 1267 reactions (HRS) on the same day as immune globulin (IG) product administrations during 2008-1268 2012. Pharmacoepidemiol Drug Safety (2013) 25:436. 1269 1270 172. Min J, Bhatt A, Aburashed R, Burton S. Cerebral venous and sinus thrombosis associated with 1271 subcutaneous immunoglobulin injection and oral contraceptive use. Neurol Sci (2012) 33:627-9. 1272 DOI: 10.1007/s10072-011-0778-y 1273 1274 173. Moltenis M, Valnet Rabier M, Rohrlich P, Kantelip J. Multiple immunoglobulin intolerance 1275 without antibody's anti-immunoglobulin A: A case report. Fundam Clin Pharmacol (2011) 1276 25(Suppl 1):72. 1277 1278 174. Thobani S, Hu E, Huynh P, Scott L. Common variable immunodeficiency: A patient with 1279 anaphylaxis to intravenous and subcutaneous immunoglobulin. J Allergy Clin Immunol (2010) 1280 125:AB143. 1281 1282 175. Moltenis M, Valnet Rabier M, Rohrlich P, Kantelip J. Multiple immunoglobulin intolerance 1283 without antibody's anti-immunoglobulin A: A case report. Fundam Clin Pharmacol (2011) 25 1284 (Suppl 1):72. 1285 1286 176. Quinti I, Soresina A, Agostini C, Spadaro G, Matucci A, Sfika I, et al. Prospective study on 1287 CVID patients with adverse reactions to intravenous or subcutaneous IgG administration. J Clin 1288 Immunol (2008) 28:263-7. doi: 10.1007/s10875-007-9169-9 1289 1290 177. Richard C, Michael B. Severe adverse reaction to subcutaneous immunoglobulin therapy in a 1291 patient with common variable immunodeficiency. Clin Exp Immunol (2013) 174:33. 1292 1293

29

178. Zenker O, Rojavin M, Baggish J, Bexon M. Evaluation of the relationship between injection 1294 site reaction rate and SCIg doses in patients with primary immunodeficiencies. J Allergy Clin 1295 Immunol (2012) 129(2 Suppl.1):AB194. 1296

Figure 1.JPEG

Figure 2.JPEG

Figure 3.JPEG

Figure 4.JPEG

Figure 5.JPEG

Figure 6.JPEG

Figure 7.JPEG

Figure 8.JPEG