<<

Immunohematology JOURNAL OF BLOOD GROUP SEROLOGY AND EDUCATION

V OLUME 20, NUMBER 3, 2004

This Issue of Immunohematology Is Supported by a Contribution From

Dedicated to Education in the Field of Blood Banking

Immunohematology JOURNAL OF BLOOD GROUP SEROLOGY AND EDUCATION VOLUME 20, NUMBER 3, 2004 CONTENTS

137 Letter to the readers Introduction to the review articles S.T.NANCE 138 Review: drug-induced immune hemolytic —the last decade G.GARRATTY 147 Review: what to do when all RBCs are incompatible—serologic aspects S.T.NANCE AND P.A.ARNDT 161 Review: transfusing incompatible RBCs—clinical aspects G. MENY 167 Review: evaluation of patients with immune L.D. PETZ 177 Case report: exacerbation of requiring multiple incompatible RBC transfusions A.M. SVENSSON,S.BUSHOR,AND M.K. FUNG 184 Delayed hemolytic transfusion reaction due to anti-Fyb caused by a primary immune response: a case study and a review of the literature H.H. KIM,T.S.PARK, S.H. OH, C.L. CHANG,E.Y.LEE,AND H.C. SON 187 Maternal alloanti-hrS—an absence of HDN R. KAKAIYA,J.CSERI,B.JOCHUM,L.GILLARD,AND S. SILBERMAN 190 193 C O M M U N I C A T I O N S Letter to the Editor-in-Chief Letter to the Editors Immunohematology to be listed in Index Medicus HAMA (Human Anti-Mouse ) do not and MEDLINE Cause False Positive Results in PAKPLUS S.G. SANDLER L.A.TIDEY,S.CHANCE,M.CLARKE,AND R.H.ASTER Reply to letter M.F.LEACH AND J.P.AUBUCHON 195 196 Letters From the Editor-in-Chief SPECIAL SECTION Ortho dedication Exerpts from the American Red Cross Reference The final 20th anniversary issue Laboratory Newsletter—1976 198 199 202 ANNOUNCEMENTS ADVERTISEMENTS INSTRUCTIONS FOR AUTHORS

EDITOR-IN-CHIEF MANAGING EDITOR Delores Mallory, MT(ASCP)SBB Mary H. McGinniss,AB, (ASCP)SBB Supply, North Carolina Bethesda, Maryland TECHNICAL EDITOR SENIOR MEDICAL EDITOR Christine Lomas-Francis, MSc Scott Murphy, MD New York, New York Philadelphia, Pennsylvania GUEST EDITOR Sandra T.Nance, MS, MT(ASCP)SBB Philadelphia, Pennsylvania ASSOCIATE MEDICAL EDITORS David Moolton, MD S. Gerald Sandler, MD Geralyn Meny, MD Ralph Vassallo, MD Philadelphia, Pennsylvania Washington, District of Columbia Philadelphia, Pennsylvania Philadelphia, Pennsylvania EDITORIAL BOARD Patricia Arndt, MT(ASCP)SBB W. John Judd, FIBMS, MIBiol Paul M. Ness, MD Los Angeles, California Ann Arbor, Michigan Baltimore, Maryland James P.AuBuchon, MD Christine Lomas-Francis, MSc Mark Popovsky, MD Lebanon, New Hampshire New York, New York Braintree, Massachusetts Geoffrey Daniels, PhD Gary Moroff, PhD Marion E. Reid, PhD, FIBMS Bristol, United Kingdom Rockville, Maryland New York, New York Richard Davey, MD Ruth Mougey, MT(ASCP)SBB Susan Rolih, MS, MT(ASCP)SBB New York, New York Carrollton, Kentucky Cincinnati, Ohio Sandra Ellisor, MS, MT(ASCP)SBB John J. Moulds, MT(ASCP)SBB David F.Stroncek, MD Anaheim, California Raritan, New Jersey Bethesda, Maryland George Garratty, PhD, FRCPath Marilyn K. Moulds, MT(ASCP)SBB Marilyn J.Telen, MD Los Angeles, California Houston, Texas Durham, North Carolina Brenda J. Grossman, MD Sandra Nance, MS, MT(ASCP)SBB St. Louis, Missouri Philadelphia, Pennsylvania

EDITORIAL ASSISTANT PRODUCTION ASSISTANT Linda Berenato Marge Manigly COPY EDITOR ELECTRONIC PUBLISHER PROOFREADER Lucy Oppenheim Paul Duquette George Aydinian

Immunohematology is published quarterly (March, June, September, and December) by the American Red Cross, National Headquarters,Washington, DC 20006. The contents are cited in the EBASE/Excerpta Medica and Elsevier BIOBASE/ Current Awareness in Biological Sciences (CABS) databases. The subscription price is $30.00 (U.S.) and $35.00 (foreign) per year. Subscriptions, Change of Address, and Extra Copies: Immunohematology, P.O. Box 40325 Philadelphia, PA 19106 Or call (215) 451-4902 Web site: www.redcross.org/pubs/immuno

Copyright 2004 by The American National Red Cross ISSN 0894-203X

LETTER TO THE READERS

Introduction to the review articles Dr. Larry Petz completes the invited review section Welcome to the third issue of 2004, celebrating with “Evaluation of Patients With Immune Hemolysis.” Immunohematology’s 20 years of publication. As with This review takes the reader through the differential the first two issues of 2004, it contains four invited diagnoses of a patient that presents with immune review articles. This issue’s focus is on diagnostic uses hemolysis and discusses the importance of the precise of immunohematology serologic testing. diagnosis in prognosis and therapy. The first review is a recurring favorite, “Drug- This third issue is a good mix of reviews that Induced Immune Hemolytic Anemia—The Last discuss serologic testing and clinical use of results for Decade,”by George Garratty, PhD. Over the years, the diagnostic and therapeutic measures. These reviews editors of Immunohematology have invited Dr. are written by authors with a great deal of experience Garratty to write review articles on drug-induced in their field. I sincerely hope you enjoy the issue as hemolytic anemia and he has kept our readers current. much as I have enjoyed being the guest editor. This review continues the tradition with another excellent review. Sandra T. Nance, MS, MT(ASCP)SBB The second and third reviews focus on what to do Member of the Editorial Board and when all units of blood are incompatible. Serologic Guest Editor of this issue, aspects are covered by Sandra Nance and Patricia American Red Cross Blood Services Arndt; these aspects include the testing often Penn-Jersey Region performed in the immunohematology reference 700 Spring Garden Street laboratory (IRL) and monocyte monolayer assays Philadelphia, PA 19123 performed as a specialty test in two IRLs in the United States. This second review also includes cases that demonstrate the points under discussion. Dr. Geralyn Meny reviews clinical responses when all units of blood are incompatible and reminds us of all the elements that should be considered when this situation arises in your facility.

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 137

Review: drug-induced immune hemolytic anemia—the last decade

G. GARRATTY

I have written three previous reviews on drug- Because of these statistics, I will be emphasizing induced immune hemolytic anemia (DIIHA) for this the cephalosporins in this review. journal.1–3 The last one was written in 1994.3 This year, I would like to review what has happened in the last Cephalosporin-Induced Immune Hemolytic decade. Anemia When Dr. Petz and I published the first edition of There are about 70 individual published case our book (Acquired Immune Hemolytic ) in reports of cephalosporin-induced immune hemolytic 1980,4 we found that there was reasonable evidence to anemia (CIIHA),8–64 but many more are contained in support that about 30 drugs could cause DIIHA. One reviews or tables without case histories65–67 (see Table drug,methyldopa,was by far the most common drug to 3). Most patients have had severe hemolytic anemia do this. Almost 70 percent of DIIHAs referred to our (HA), often with intravascular , and 40 percent laboratory in the 1970s were associated with methyl- were associated with fatal HA. It is not known if this is dopa. Antibodies associated with methyldopa were the tip of the iceberg and there are many more cases drug-independent and the patients showed all the serologic and hematologic characteristics of “warm of milder HA or positive DATs that are not reported; type” autoimmune hemolytic anemia (WAIHA). the same questions apply to cephalosporin-induced Twenty-three percent of the DIIHAs were associated thrombocytopenia. Tables 4 and 5 summarize the with high-dose IV penicillin therapy; only about 10 clinical and serologic findings associated with percent of DIIHAs were associated with other drugs cefotetan- and ceftriaxone-induced immune HA. It (e.g., quinine, rifampicin, and hydrochlorothiazide). should be emphasized that cefotetan antibodies always In the next 20 years, methyldopa and high dose IV react with cefotetan-coated RBCs and almost always penicillin were used less and less; we have not seen a react with untreated RBCs in the presence of cefotetan case of DIIHA associated with these two drugs for (“immune complex” method), and about one-third will many years. By 1994 about 71 drugs had been react with RBCs without the presence of drug (i.e.,will implicated in DIIHA.5 Recently published results appear to be autoantibodies). The latter findings can reflect a changing picture in the spectrum of DIIHA in lead to problems in the blood transfusion service. If a the last 25 years.6,7 There are now approximately 100 patient receives cefotetan prophylactically for surgery, drugs associated with DIIHA (see Table 1), and receives a blood transfusion during or after surgery,and methyldopa and penicillin have been replaced by a then develops HA 7 to 10 days afterwards, a delayed single group of drugs, the cephalosporins (93% of hemolytic transfusion reaction is often suspected. The cases), with cefotetan alone accounting for 83 percent hematologic findings can also mimic AIHA. If the HA is of the DIIHAs we have encountered in the last 10 due to cefotetan,the DAT will be positive (although we years. Table 2 shows the drugs causing DIIHA that we have reported one case where the DAT was negative).68 have encountered in the past 26 years (1978–2003). Sometimes the serum will react with all untreated Methyldopa is probably underrepresented as, by 1978, RBCs, mimicking an alloantibody to a high-frequency cases of autoimmune hemolytic anemia (AIHA) in antigen, or a mixture of alloantibodies or autoantibody, patients taking methyldopa were not usually sent to and many hours may be wasted investigating these specialist laboratories such as ours for investigation. possibilities. If there is a history of cefotetan

138 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 Review: drug-induced hemolytic anemia

Table 1. Drugs causing immune hemolytic anemia and/or positive DATs* aceclofenac diethylstilbestrol nomifensine acetaminophen/paracetamol diglycoaldehyde (INOX) oxaliplatin aminopyrine/pyramidon diphenylhydantoin p-aminosalicylic acid amphotericin B dipyrone penicillin G ampicillin erythromycin phenacetin antazoline etodolac phenytoin apazone/azapropazone fenfluramine piperacillin apronalide fenoprofen probenecid butizide fludarabine procainamide carbenicillin fluorescein propyphenazone carbimazole fluoroquinolone quinidine carboplatin fluorouracil (5-FU) quinine carbromal furosemide ranitidine catergen/cyanidanol glafenine rifampicin cefamandole hydralazine rituximab cefazolin hydrochlorothiazide sodium pentothal/thiopental cefixime 9-hydroxy-methyl-ellipticinium stibophen cefotaxime ibuprofen streptomycin cefotetan indene derivatives (e.g., sulindac) sulbactam sodium (e.g., in Unasyn) cefoxitin insulin sulindac ceftazidime interferon sulfonamides ceftizoxime interleukin-2 sulfonylurea derivatives (e.g., chlorpropamide) ceftriaxone isoniazid suprofen cephalexin latamoxef suramin cephalordine levodopa tazobactam (e.g., in Zosyn) cephalothin mefenamic acid teicoplanin chaparral mefloquine temafloxacin chlordiazepoxide melphalan teniposide chlorinated hydrocarbons mephenytoin tetracycline chlorpromazine methoin ticarcillin chlorpropamide 6-mercaptopurine tolbutamide cianidanol methadone tolmetin cisplatin methicillin triamterene cladribine methotrexate trimellitic anhydride clavulanate potassium (e.g.,Timentin) methyldopa zomepirac cyclofenil methysergide diclofenac nafcillin

*Drugs in publications containing reasonable evidence for an immune etiology were the only drugs included (many more are in the literature).

Table 2. Drugs associated with DIIHA investigated at American Red Table 3. Cephalosporins reported to cause immune hemolytic anemia Cross Blood Services, Los Angeles, in the last 26 yrs. (up to 2003) (1978–2003) Drug Number of case reports References Drug Number of patients cephalothin 5 8–11 cefotetan 74 cefazolin 1 11 ceftriaxone 12 cephalexin 2 12, 13 piperacillin 5 cefamandole 1 14 tazobactam 5 cefoxitin 2 15, 16 clavulanate 3 cefotaxime 3 17–19 ceftriaxone 19 20–37 fludarabine 2 cefotetan 35* 37–59 penicillin 2 ceftizoxime 4 60–62 probenecid 2 cefixime 1 19 rifampicin 2 ceftazidime 2 63, 64 cefotaxime 1 sulbactam 1 Total 75 ticarcillin 1 *Many other cases are reported, without individual case reports, in references 65 (43 mefloquine 1 cases) and 66 (85 cases) cefoxitin 1 chlorpropamide 1 administration, I recommend testing an eluate from the nafcillin 1 patient’s RBCs against untreated and cefotetan-coated phenacetin 1 RBCs to help with the differential diagnosis. If the procainamide 1 eluate is reactive with cefotetan-treated RBCs and not erythromycin 1 tolmetin 1 the same untreated RBCs, the diagnosis is obvious. oxaliplatin 1 Unfortunately, in about 15 percent of cases we have Total 119 studied, the eluate also reacted with untreated RBCs

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 139 G. GARRATTY

Table 4. Clinical* and serologic** findings associated with cefotetan- pH does not reduce sensitivity but does reduce induced IHA nonspecific uptake of leading to falsely • Approx. 80 percent of patients received cefotetan for surgery; usually a single dose of 2 g was used. positive antihuman globulin (AHG) tests. Some • A history of previous cefotetan therapy was not common. investigators have suggested that drugs can be • HA was obvious in less than 1 day to 13 days after receiving cefotetan. solubilized more efficiently in 1% albumin.70 We do Only two patients had HA in < 1 day; the mean of the other 29 was 9 days. not advise this when testing for cefotetan • Patients’ nadir Hb after receiving cefotetan = 2.6 g/dL (mean = 4.8 g/dL). antibodies, as albumin can cause reduced binding • Most patients had signs of intravascular lysis (hemoglobinemia/ of drug to drug-treated RBCs.71 hemoglobinuria). 2. Most individuals, including healthy donors, have • Fatal HA and renal failure occurred in 19 percent of patients. • Patients always had a positive DAT: antibodies in their sera that can directly agglutinate – 100% had RBC-bound IgG cefotetan-coated RBCs.72 In addition,almost all sera – 86% had RBC-bound C3 – 44% had RBC-bound IgA (including those from healthy individuals) will – 7.4% had RBC-bound IgM yield positive IATs with cefotetan-treated RBCs. • All sera agglutinated cefotetan-treated RBCs (median titer 512) and When testing a patient’s serum with cefotetan- reacted by IAT (median titer 16,000); all normal sera also reacted with 36 the cefotetan-treated RBCs, but were nonreactive when diluted ≥ 1 in coated RBCs, dilute the serum 1 in 100 in saline. 100. This will avoid agglutination and nonspecific • All but one of the sera reacted with untreated RBCs in the presence of protein uptake onto cefotetan-treated RBCs; cefotetan (“immune complex” mechanism). • 33 percent and 40 percent of sera reacted with RBCs without drug clinically significant antibodies always have titers being present, with saline-suspended RBCs or in the presence of PEG, > 100 (e.g., in the thousands). Some texts still 65 respectively. recommend a dilution of 1 in 20, which is what we *Clincal data from references 38–40, 42–49, 50–53, 56, 57 ** Serologic data from reference 65 recommended when using cephalothin-coated RBCs. We have found that 1 in 20 is sufficient to exclude nonspecific adsorption of by Table 5. Clinical and serologic findings associated with ceftriaxone- induced IHA cefotetan-treated RBCs, but is not sufficient to • HA is more acute and severe in children compared to adults. In exclude agglutination of cefotetan-treated RBCs by children, HA started 5 minutes to 7 days after receiving drug; for six about 10 percent of sera (even from healthy children there was a mean of 27 minutes and for three children a mean 72 of 6 days.22,26–29,31,34,37 In adults, HA started 30 minutes to 34 days. donors). One patient started after 30 minutes; the mean for nine others was 3. The lesson to be learned by the above is: never 20,21,27,28,30,32,33,35,36 9 days. report cefotetan antibodies to be present based on • Fatal HA occurred in 67 percent of 9 children and 30 percent of ten adults. reactions of undiluted sera. As with penicillin- • There is a history of previous ceftriaxone therapy. induced immune HA,the diagnosis should be based • Positive DATs associated with complement are present in all cases and on an eluate from the patient’s DAT-positive RBCs additional IgG is present in 75 percent; none had detectable RBC-bound reacting with cefotetan-coated RBCs, but not 65 IgA or IgM. untreated RBCs; unlike penicillin, the cefotetan • Antibodies are detected only by immune complex method65 (serum + drug + RBCs). Some patients’ sera only react with a metabolite of in the eluate may sometimes react with ceftriaxone (ex vivo antigen).29,32,35 untreated RBCs (see reference 65). Another problem is that the last wash of the patient’s RBCs may be reactive.73 The presence of a high titer (> (Arndt, Leger, Garratty, unpublished observations). The 100) cefotetan antibody in the serum confirms the diagnosis may then rest on the presence of a high-titer diagnosis. (> 100) cefotetan antibody in the patient’s serum. 4. Ceftriaxone-coated RBCs cannot be made, thus the Some illustrative case histories associated with such serologic diagnosis is based on the results of the problems have been published in this journal.36 “immune-complex” method. Nonspecific ad- sorption of proteins is not a problem by this Technical hints when investigating CIIHA method, thus undiluted sera can be used. 1. When preparing cefotetan-coated RBCs, use a Sometimes, positive reactions are only obtained by buffer with a pH of 6 to 7 (or normal saline) instead using -treated RBCs, or metabolites of the of the high pH buffer (pH 8 to 10) used for drug (e.g., in the presence of urine from a patient preparation of penicillin-coated RBCs.69 The lower taking ceftriaxone).29,32,35

140 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 Review: drug-induced hemolytic anemia

DIIHA Associated With Nonimmunologic Drugs belonging to the platinum family (e.g., Adsorption of Proteins Onto RBCs cisplatin, carboplatin, and oxaliplatin) have been RBCs treated with the first-generation cephalo- associated with positive DATs and HA.81–92 Some sporin, cephalothin, were found to adsorb many investigators81 have suggested that cisplatin causes proteins nonimmunologically when incubated in vitro nonimmunologic adsorption of protein and that the with normal plasma/sera.74 Such adsorbed proteins led HA is coincidental; others believe the drug can cause to a positive AHG test, but it was thought that this was DIIHA. We believe that some patients can develop clinically insignificant, and indeed there were only five antibodies to the drug and that the DIIHA may occur cases of DIIHA due to cephalothin reported in more because of this, regardless of nonimmunologic protein than 30 years of use.8–11 Some other drugs have been adsorption. We have described a patient who had found to show a similar phenomenon (see Table 6), but antibodies to oxaliplatin but no HA.92 some of these drugs have been thought to cause DIIHA Nevertheless, I now believe that nonimmunologic more commonly than cephalothin. adsorption of protein may lead to HA. This is mainly based on work we have performed on another family Table 6. Drugs associated with nonimmunologic adsorption of proteins onto RBCs of drugs, the beta lactamase inhibitors. Examples of these commonly used drugs are sulbactam (contained cephalosporins cisplatin/oxaliplatin/carboplatin in Unasyn), clavulanate (contained in Augmentin and diglycoaldehyde (INOX) Timentin), and tazobactam (contained in Zosyn). They suramin are used together with the beta lactam antibiotics sulbactam (contained in Unasyn) clavulanate (contained in Augmentin and Timentin) ampicillin, ticarcillin, and piperacillin, respectively. tazobactam (contained in Zosyn) Lutz and Dzik93 reported that 39 percent of patients receiving Unasyn developed weakly positive DATs but Causes for the uptake of protein are not always no HA. We showed later that RBCs treated in vitro with clear. Garratty and Petz75 suggested that cephalothin the beta lactamase inhibitors contained in Unasyn, changed the RBC membrane in a way that created sites Augmentin,Timentin,and Zosyn would adsorb proteins for protein binding. Later this was challenged by nonimmunologically, leading to positive IATs, and that Branch and Petz,76 who suggested that no membrane this may be the cause of positive DATs associated with change occurred and that protein attached to free such drugs. It should be mentioned that patients may protein binding sites on the cephalothin that had make antibodies to the antibiotics also present in the covalently bonded to proteins on the RBC membrane. drug (e.g.,ampicillin,piperacillin),and these antibodies Garratty and Leger77 were stimulated by an old finding, can cause positive AHG tests and HA. Garratty and that cefotetan-treated RBCs were “PNH-like”(e.g., had a Arndt94,95 suggested that HA could also occur in positive acidified serum test).78 To reexamine the issue, patients who have no antibodies to the antibiotics and Garratty and Leger77 confirmed these old findings by that this may be associated with nonimmunologic showing, using flow cytometry, that cephalothin- and adsorption of protein. This suggestion was supported cefotetan-treated RBCs had markedly decreased by case histories and in vitro experiments where it was amounts of CD55 and CD58 on the RBC membrane; shown that RBCs treated with the beta lactamase CD59 was only slightly decreased. Thus, the results inhibitors, washed, and incubated in normal plasma, were different from the changes associated with PNH washed, and then added to a monocyte monolayer but confirmed the original hypothesis of Garratty and yielded results suggesting that the RBCs would have 75 Petz that cephalosporins can modify the RBC shortened survival.95 Broadberry et al.96 described a membrane. Whether this is the cause of the DIIHA associated with tazobactam;the HA was thought nonimmunologic uptake of protein remains to be to be due to nonimmunologic adsorption of protein. proved. Unfortunately, we cannot prove in the laboratory Other drugs have been found to cause non- that this nonimmunologic adsorption of proteins is immunologic adsorption of proteins onto RBCs (Table causing the HA in a particular patient; one can only 6). Some drugs containing aldehyde groups can bind to suggest it to the physicians and refer them to the RBC proteins and adsorb proteins from the plasma, appropriate literature (e.g., references 6 and 94–96). leading to positive AHG tests, but HA has not been associated with such drugs.79,80

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 141 G. GARRATTY

Mechanisms of DIIHA Antibody to Drug There are two well-accepted mechanisms for DIIHA, namely, that individuals can produce either drug-dependent antibodies or drug-independent Antibody to antibodies (Table 7). The latter can be classic (Mainly) Membrane Drug Components autoantibodies in that they do not require drug to be Antibody to Drug and Membrane Table 7. Drugs that have been reported to induce RBC drug-independent Components antibodies (i.e., autoantibodies)* Group 1 Group 1a Group 2 cladribine catergen azapropazone fludarabine chaparral carbimazole levodopa chlordiazepoxide ceftazidime Red mefenamic acid cianidanol cefoxitin methyldopa cyclofenil cefotetan Fig. 1. Proposed unifying theory of drug-induced antibody reactions.97 procainamide diphenylhydantoin cefotaxime The thicker, darker lines represent antigen-binding sites on the fenfluramine chlorinated F(ab) region of the drug-induced antibody. Drugs (haptens) bind ibuprofen hydrocarbons loosely or firmly to cell membranes, and antibodies may be made interleukin-2 diclofenac to the drug (producing in vitro reactions typical of a drug methoin glafenine adsorption [penicillin-type] reaction; the membrane compo- methysergide latamoxef nents, or mainly membrane components (producing in vitro rituximab nomifensine reactions typical of autoantibody); or part-drug, part-membrane phenacetin components (producing an in vitro reaction typical of the so- streptomycin called immune-complex mechanism). teniposide tolmetin zomepirac Drug-dependent antibodies can be of two types. One type of antibody can be inhibited by the drug *Drugs in groups 1 and 1a have been reported to induce drug-dependent antibodies only. More evidence is needed to prove that drugs in group 1a really can induce RBC (hapten inhibition) and reacts only with washed, drug- autoantibodies. Drugs in group 2 induce drug-independent antibodies together with coated RBCs (prototype drug is penicillin). The other antibodies reacting by different mechanisms. type, which relates to most drugs, is not inhibited by the drug and reacts with untreated RBCs in the present for their reaction with RBCs in vitro or in vivo; presence of the drug. We still do not know why. There drug is only needed to initiate the production of have been two main suggestions: antibodies. It is still unclear how this occurs, but the • The drug antibody combines with the drug, most popular concept is that certain drugs given to forming immune complexes which attach to the certain patients somehow affect the immune system to RBC (transiently) and activate complement. produce pathogenic autoantibodies, as do certain • The drug changes the RBC membrane, producing in certain patients. The mechanisms may a new antigen (“neoantigen”) as a new epitope. turn out to be similar. The AIHA often continues for This neoantigen may be a chemically modified some time after the drug is discontinued. Some drug or part drug, part RBC antibodies (e.g., cefotetan) sometimes react with RBCs membrane protein (“compound” epitope). without drug being present, but I do not think that The latter part of the latter concept is the basis of these are “classic” autoantibodies (in contrast to those the unifying hypothesis originally suggested by associated with methyldopa). Drug-independent Habibi,98 and supported by Mueller-Eckhardt and antibodies (e.g., cefotetan antibodies) are never seen Salama,99 which I feel has much merit. Unfortunately, without the presence of drug-dependent antibodies like other concepts, it has some failings (these are and when the drug is stopped, the “autoantibodies” discussed in references 5, 6, and 99). Although the become weaker, eventually disappearing (see Table 7). immune complex hypothesis is out of favor, the clinical I believe that these antibodies are not produced findings and some serologic data fit much better with because of the drug’s effect on the immune system,but the “immune complex”hypothesis. Shulman and Reid100 rather are part of the spectrum of antibodies produced tried to take some of the best of both hypotheses, and as a result of the immune response to a hapten (e.g., suggest some explanations for the discrepancies, but antibodies to the drug, carrier [RBC protein], and drug personally I do not think that, in 2004, we have any + carrier).97 See Figure 1. better explanations that we did in 1994!

142 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

Review: drug-induced hemolytic anemia

References 16. DeTorres OH. Hemolytic anemia and pancytopenia 1. Garratty G. Drug induced immune hemolytic induced by cefoxitin. Drug Intell Clin Pharm anemia and/or positive direct antiglobulin tests. 1983;17:816-8. Immunohematology 1985;2:1-8. 17. Shulman IA, Arndt PA, McGehee W. Cefotaxime- 2. Garratty G. Current viewpoints on mechanisms induced immune hemolytic anemia due to causing drug-induced immune hemolytic anemia antibodies reacting in vitro by more than one and/or positive direct antiglobulin tests. mechanism.Transfusion 1990;30:263-6. Immunohematology 1989;5:97-106. 18. Salama A, Göttsche B, Schleffer T, et al. ‘Immune 3. Garratty G. Immune hemolytic anemia and/or complex’ mediated intravascular hemolysis due positive direct antiglobulin tests caused by drugs. to IgM cephalosporin dependent antibody. Immunohematology 1994;10:41-50. Transfusion 1987;27:460-3. 4. Petz LD, Garratty G. Acquired immune hemolytic 19. Malaponte G, Arcidiacono C, Mazzarino C, et al. anemias. New York: Churchill Livingstone, 1980. Cephalosporin-induced hemolytic anemia in a 5. Garratty, G. Drug-induced immune hemolytic an- Sicilian child. Hematology 2000;5:327-34. emia. In: Garratty G, ed. Immunobiology of trans- 20. Garratty G, Postoway N, Schwellenbach J, et al. A fusion medicine. New York: Dekker, 1994:523-51. fatal case of ceftriaxone (Rocephin)-induced 6. Petz LD, Garratty G. Immune hemolytic anemias. 2nd ed. Philadelphia: Churchill Livingstone,2004: hemolytic anemia associated with intravascular 261-317. immune hemolysis. Transfusion 1991;31:176-9. 7. Garratty G, Arndt P, Leger R. The changing 21. Lo G, Higginbottom P. Ceftriaxone induced spectrum of drug-induced immune hemolytic hemolytic anemia (abstract). Transfusion 1993;33: anemia over the last 25 years (abstract). Blood (Suppl):25S. 2003;102:560a. 22. Bernini JC, Mustafa MM, Sutor LJ, et al. Fatal 8.Gralnick HR, McGinniss M, Elton W, et al. hemolysis induced by ceftriaxone in a child with Hemolytic anemia associated with cephalothin. sickle cell anemia. J Pediatr 1995;126:813-5. JAMA 1971; 217:1193-7. 23. Lascari AD, Amyot K. Fatal hemolysis caused by 9. Jeannet M, Bloch A, Dayer JM, et al. Cephalothin- ceftriaxone. J Pediatr 1995;126:816-7. induced immune hemolytic anemia. Acta 24. Borgna-Pignatti C, Bezzi TM, Reverberi R. Fatal Haematol 1976;55:109-17. ceftriaxone-induced hemolysis in a child with 10. Rubin RN, Burka ER. Anti-cephalothin antibody acquired immunodeficiency syndrome. Pediatr and Coombs’-positive hemolytic anemia. Ann Infect Dis J 1995;14:1116-7. Intern Med 1977; 86:64-5. 25. Scimeca PG, Weinblatt ME, Boxer R. Hemolysis 11. Moake JL, Butler CF,Hewell GM, et al. Hemolysis after treatment with ceftriaxone (letter). J Pediatr induced by cefazolin and cephalothin in a patient 1996;128:163. with penicillin sensitivity. Transfusion 1978;18: 26. Moallem HJ, Garratty G, Wakeham M, et al. 369-73. Ceftriaxone-related fatal hemolysis in an 12. Forbes CD, Mitchell R, Craig JA, et al. Acute intra- adolescent with perinatally acquired human vascular haemolysis associated with cephalexin immunodeficiency virus . J Pediatr 1998; therapy. Postgrad Med J 1972;48:186-8. 133:279-81. 13. Manoharan A, Kot T. Cephalexin-induced haemo- lytic anaemia (letter). Med J Aust 1987;147:202. 27. Longo F, Hastier P, Buckley MJM, et al. Acute 14.Branch DR, Berkowitz LR, Becker RL, et al. hepatitis, autoimmune hemolytic anemia, and Extravascular hemolysis following the adminis- erythroblastocytopenia induced by ceftriaxone. tration of cefamandole. Am J Hematol 1985;18: Am J Gastroenterol 1998;93:836-7. 213-9. 28. Maraspin V, Lotric-Furlan S, Strle F. Ceftriaxone- 15. Toy E, Nesbitt R, Savastano G, et al. Warm associated hemolysis. Wien Klin Wochenschr autoantibody following plasma apheresis, compli- 1999;111(9):368-70. cated by acute intravascular hemolysis associated 29. Meyer O, Hackstein H, Hoppe B, et al. Fatal with cefoxitin-dependent antibody resulting in immune haemolysis due to a degradation product fatality (abstract).Transfusion 1989;29(Suppl):51S. of ceftriaxone. Br J Haematol 1999;105:1084-5.

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 143 G. GARRATTY

30. Punar M, Özsü H, Eraksoy H, et al. An adult case of 42. Ehmann WC. Cephalosporin-induced hemolysis: a fatal hemolysis induced by ceftriaxone. Clin case report and review of the literature. Am J Microbiol Infect 1999;5:585-6. Hematol 1992;40:121-5. 31. Viner Y, Hashkes PJ, Yakubova R, et al. Severe 43.Chenoweth CE, Judd WJ, Steiner EA, et al. hemolysis induced by ceftriaxone in a child with Cefotetan-induced immune hemolytic anemia. sickle cell anemia. Pediatr Infect Dis J 2000;19(1): Clin Infect Dis 1992;15:863-5. 83-5. 44. Wagner BKJ, Heaton AH, Flink JR. Cefotetan 32. Seltsam A, Salama A. Ceftriaxone-induced immune disodium-induced hemolytic anemia. Ann haemolysis: two case reports and a concise review Pharmacother 1992;26:199-200. of the literature. Intensive Care Med 2000;26: 45. Dhawan M, Kiss JE, DiAddezzio NA, et al. Fatal 1390-4. cefotetan-induced immune hemolysis (abstract). 33.Falezza GC, Piccoli PL, Franchini M, et al. Blood 1993;82:582a. Ceftriaxone-induced hemolysis in an adult (letter). 46. Eckrich RJ, Fox S, Mallory D. Cefotetan-induced Transfusion 2000;40:1543-5. immune hemolytic anemia due to the drug- 34. Citak A, Garratty G, Ücsel R, et al. Ceftriaxone- adsorption mechanism. Immunohematology induced haemolytic anaemia in a child with no 1994;10:51-4. immune deficiency or haematological disease. 47. Peano GM, Menardi G, Quaranta L, et al. A rapidly J Pediatr Child Health 2002;38:209-10. fatal case of immune haemolytic anaemia due to 35. Kim S, Song KS, Kim HO, et al. Ceftriaxone cefotetan.Vox Sang 1994;66:84-5. induced immune hemolytic anemia: detection of 48. Ogburn JR, Knauss MA,Thapar K, et al. Cefotetan- induced immune hemolytic anemia (IHA) drug-dependent antibody by ex-vivo antigen in resulting from both drug adsorption and immune urine. Yonsei Med J 2002;43:391-4. complex mechanisms (abstract). Transfusion 36. Arndt P. Practical aspects of investigating drug- 1994;34(Suppl):27S. induced immune hemolytic anemia due to 49. Mohammed S, Knoll S, vanAmburg A III, et al. cefotetan or ceftriaxone—a case study approach. Cefotetan-induced hemolytic anemia causing Immunohematology 2002;18:27-32. severe hypophosphatemia. Am J Hematol 1994; 37. Eastlund T,Mulrooney D,Neglia J,et al. Self-limited 46:369-70. immune hemolysis in a child after six days of 50. Garratty G, Leger RM, Arndt PA. Severe immune ceftriaxone therapy (abstract). Transfusion 2002; hemolytic anemia associated with prophylactic 42(Suppl):96S. use of cefotetan in obstetric and gynecologic 38. Garratty G, Nance S, Lloyd M, et al. Fatal immune procedures. Am J Obstet Gynecol 1999;9:337-42. hemolytic anemia due to cefotetan. Transfusion 51. Badon SJ, Cable RG. Hemolysis due to cefotetan 1992;32:269-71. (abstract).Transfusion 1999;39(Suppl):42S. 39.Weitekamp LA, Johnson ST, Fueger JT, et al. 52. Johnson ST, Fueger JT, Gottschall JL. Cefotetan- Cefotetan-dependent immune hemolytic anemia dependent antibody cross-reacting with ceftri- due to a single antibody reacting with both drug axone treated RBCs (abstract). Transfusion 1999; coated and untreated red cells in the presence 39(Suppl):81S. of drug. Book of Abstracts. ISBT/AABB Joint 53. Stroncek D, Proctor JL, Johnson J. Drug-induced Congress. Arlington, VA: American Association of hemolysis: cefotetan-dependent hemolytic anemia Blood Banks 1990:33. mimicking an acute intravascular immune 40. Wojcicki RE, Larson CJ, Pope ME, et al. Acute transfusion reaction. Am J Hematol 2000;64:67-70. hemolytic anemia during cefotetan therapy. 54. Naylor CS,Steele L,Hsi LR,et al. Cefotetan-induced Book of Abstracts. ISBT/AABB Joint Congress. hemolysis associated with antibiotic prophylaxis Arlington,VA: American Association of Blood for cesarean delivery. Am J Obstet Gynecol 2000; Banks 1990:33. 182:1427-8. 41. Gallagher MT, Schergen AK, Sokol-Anderson ML, 55. Moes GS, MacPherson BR. Cefotetan-induced et al. Severe immune mediated hemolytic anemia hemolytic anemia: a case report and review of the secondary to treatment with cefotetan. literature. Arch Pathol Lab Med 2000;124(a): Transfusion 1992;32:266-8. 1344-6.

144 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

Review: drug-induced hemolytic anemia

56. Marques MB, Carr KD, Brumfield CG, et al. A 68. Leger RM,Arndt PA, Garratty G. A negative or very pregnant patient with sickle cell disease and weakly positive DAT should not exclude the cefotetan-induced immune hemolysis. Lab Med investigation of drug-induced immune hemolytic 2000;31(10):541-3. anemia. Transfusion 2003;43:1640-1. 57. Ray EK, Warkentin TE, O’Hoski PL, et al. Delayed 69.Petz LD, Branch DR. Drug-induced immune onset of life-threatening immune hemolysis after hemolytic anemia. In: Chaplin H Jr., ed. Immune perioperative antimicrobial prophylaxis with hemolytic anemias. New York: Churchill cefotetan. Can J Surg 2000;43:461-2. Livingstone, 1985:47-94. 58. Chai L,Pomper GJ,Ross RL,et al. Severe hemolytic 70. Osbourne SE, Johnson ST, Weitekamp LE, et al. anemia and liver damage induced by prophylactic Enhanced detection of drug-dependent antibodies use of cefotetan (abstract). Transfusion 2001;41 reacting with untreated red blood cells in the (Suppl 1):58S. presence of drug (abstract). Transfusion 1993; 59. Afenyi-Annan AN, Judd WJ. Cefotetan induced 34(Suppl):20S immune-mediated hemolysis complicated by 71. Arndt P,Garratty G. Use of albumin solutions for thrombocytopenia: alloimmune or thrombotic solubilizing certain drugs can decrease binding of (abstract)? Transfusion 2002;52(Suppl):46S. drugs to RBCs (abstract). Transfusion 2002;42 60. Shammo JM, Calhoun B, Mauer AM, et al. First two (Suppl):105S. cases of immune hemolytic anemia associated 72. Arndt PA, Garratty G. Is severe immune hemolytic with ceftizoxime. Transfusion 1999;39:838-44. anemia, following a single dose of cefotetan, 61. Endoh T,Yagihashi A, Sasaki M, et al. Ceftizoxime- associated with the presence of “naturally- induced hemolysis due to immune complexes: occurring” anti-cefotetan? (abstract). Transfusion case report and the epitope responsible for 2001;41(Suppl):24S. immune complex-mediated hemolysis. Transfusion 73. Arndt PA, Leger RM, Garratty G. Reactivity of “last 1999;39: 306-9. wash” elution controls in investigations of 62. Calhoun BW, Junsanto T, Donoghue MT, et al. cefotetan antibodies (abstract). Transfusion 1999; Ceftizoxime-induced hemolysis secondary to 39:(Suppl):47S. combined drug adsorption and immune-complex 74. Spath P, Garratty G, Petz LD. Studies on the mechanisms.Transfusion 2001;41:893-7. immune response to penicillin and cephalothin in 63. Chambers LA, Donovan LM, Kruskall MS. humans. II. Immunohematologic reactions to Ceftazidime-induced hemolysis in a patient with cephalothin administration. J Immunol 1971; drug-dependent antibodies reactive by immune 107:860-9. complex and drug adsorption mechanisms. Am J 75.Garratty G, Petz LD. Drug-induced immune Clin Pathol 1991;95:393-6. hemolytic anemia. Am J Med 1975;58:398-407. 64. Fueger JT,Bell JA, Gottschall JL, et al. Ceftazidime- 76. Branch DR, Sy Siok Hian AL, Petz LD. Mechanism dependent antibody reacting with untreated red of nonimmunologic adsorption of proteins using cells in the presence of drug (abstract). cephalothin-coated red cells (abstract). Transfusion 2001;41(Suppl 1):104S. Transfusion 1985;24:415. 65. Arndt PA, Leger RM, Garratty G. Serology of 77. Garratty G, Leger R. Red cell membrane proteins antibodies to second- and third-generation (CD55 and CD58) are modified following cephalosporins associated with immune hemo- treatment of RBCs with cephalosporins (abstract). lytic anemia and/or positive direct antiglobulin Blood 1995;86:68a. tests. Transfusion 1999;39:1239-46. 78. Sirchia G, Murcuriali F, Ferrone S. Cephalothin- 66.Viraraghavan R, Chakravarty AG, Soreth J. treated normal red cells: a new type of PNH-like Cefotetan-induced haemolytic anaemia. A review cells. Experientia 1968;24:495-6. of 85 cases. Adverse Drug React Toxicol Rev 79. Jamin D, Demers J, Shulman I, et al.An explanation 2002;21:101-7. for nonimmunologic adsorption of proteins onto 67. Bateman ST, Hu E, Lane C, et al. Antibody to red blood cells. Blood 1986;67:993-6. ceftriaxone in HIV pediatric patients and potential 80. Jamin D, Shulman I, Lam HT, et al. Production of a implications for RBC hemolysis (abstract). Blood positive direct antiglobulin test due to Suramin. 2002;100(11):48b. Arch Pathol Lab Med 1988;112:898-900.

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 145 G. GARRATTY

81. Zeger G, Smith L, McQuiston D, et al. Cisplatin- 94. Garratty G, Arndt PA. Positive direct antiglobulin induced nonimmunologic adsorption of immuno- tests and haemolytic anaemia following therapy globulin by red cells.Transfusion 1988; 28:493-5. with beta-lactamase inhibitor containing drugs 82. Getaz EP, Beckley S, Fitzpatrick J, et al. Cisplatin- may be associated with nonimmunologic induced hemolysis. N Engl J Med 1980;302:334-5. adsorption of protein onto red blood cells. Br J 83.Levi JA, Aroney RS, Dalley DN. Haemolytic Haematol 1998;100:777-83. anaemia after cisplatin treatment. Br Med J (Clin 95. Arndt PA, Leger RM, Garratty G. Positive direct Res Ed) 1981;282:2003-4. antiglobulin tests and haemolytic anaemia 84. Nguyen BV,Jaffe N, Lichtiger B. Cisplatin-induced following therapy with the beta-lactamase anemia. Cancer Treat Rep 1981; 65:1121. inhibitor, tazobactam, may also be associated with 85. Cinollo G, Dini G, Franchini E, et al. Positive direct non-immunologic adsorption of protein onto red antiglobulin test in a pediatric patient following blood cells (letter). Vox Sang 2003;85:53. high-dose cisplatin. Cancer Chemother Pharmacol 96.Broadberry RE, Farren TW, Kohler JA, et al. 1988;21:85-6. Tazobactum-induced haemolytic anaemia possibly 86. Weber JC, Couppie P, Maloisel F, et al. Anémie caused by non-immunological adsorption of IgG hémolytique au cisdiamino-dichloroplatinum. La onto patient’s red cells. Transfus Med 2004; Press Médicale 1990;19:526-7. 14:53-7. 87.Marani TM, Trich MB, Armstrong KS, et al. 97. Garratty G. Target antigens for red-cell-bound Carboplatin-induced immune hemolytic anemia. autoantibodies. In: Nance SJ, ed. Clinical and Transfusion 1996;36:1016-8. basic science aspects of immunohematology. 88. Desrame J, Broustet H, de Talilly PD, et al. Arlington, VA: American Association of Blood Oxaliplatin-induced haemolytic anaemia. Lancet Banks, 1991:33-72. 1999;354:1179-80. 98. Habibi B. Drug induced 89. Garufi C, Vaglio S, Brienza S, et al. Immuno- autoantibodies co-developed with drug specific hemolytic anemia following oxaliplatin adminis- antibodies causing haemolytic anaemias. Br J tration (letter). Ann Oncol 2000;11:497. Haematol 1985;61:139-43. 90. Earle CC, Chen WY, Ryan DP et al. Oxaliplatin- 99. Mueller-Eckhardt C, Salama A. Drug-induced induced Evan’s syndrome (letter). Br J Cancer immune cytopenias: a unifying pathogenetic 2001;84:441. concept with special emphasis on the role of drug 91. Sørbye H, Bruserud Ø, Dahl O. Oxaliplatin- metabolites.Transfus Med Rev 1990;4:69-77. induced haematological emergency with an 100. Shulman NR, Reid DM. Mechanisms of drug- immediate severe thrombocytopenia and induced immunologically mediated cytopenias. haemolysis. Acta Oncol 2001;40:882-3. Transfus Med Rev 1993;VII:215-29. 92. Arndt P, Garratty G. Positive direct antiglobulin tests associated with oxaliplatin can be caused by George Garratty, PhD, FRCPath, American Red Cross antibody and/or nonimmunological protein Blood Services, Southern California Region, 1130 S. adsorption (abstract). Transfusion 2003 43:102a. Vermont Avenue, Los Angeles, CA 90006. e-mail:ggar- 93. Lutz P, Dzik W. Very high incidence of a positive [email protected]. direct antiglobulin test (+DAT) in patients receiving Unasyn® (abstract). Transfusion 1992; 32(Suppl):23S.

Phone, Fax, and Internet Information: If you have any questions concerning Immunohematology, Journal of Blood Group Serology and Education, or the Immunohematology Methods and Procedures manual,contact us by e-mail at [email protected] information concerning the National Reference Laboratory for Blood Group Serology, including the American Rare Donor Program, please contact Sandra Nance, by phone at (215) 451-4362, by fax at (215) 451-2538, or by e-mail at [email protected]

146 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 Review: what to do when all RBCs are incompatible—serologic aspects

S.T.NANCE AND P.A.ARNDT

A serologic workup to investigate the presence of facility. The patient’s sample should be tested against a irregular antibodies in a patient’s sample includes panel of reagent RBCs, either at the facility or at an testing the patient’s serum (or plasma) against a panel immunohematology reference laboratory (IRL). Since of reagent RBCs and the patient’s autologous RBCs. If this was a three-cell screen and all RBCs tested were all panel RBCs and the autologous RBCs are equally reactive, the chance of this being a simple antibody is reactive, then the most likely conclusion is that the low and reactions of the three cells on the gel card patient’s serum contains a warm autoantibody and/or a should be reviewed to see if there are differences in the cold autoantibody. If all panel RBCs are reactive and appearance of the reactions with each of the three RBC the autologous RBCs are nonreactive, then the patient suspensions. If there are differences, then multiple has either multiple alloantibodies and/or an antibody antibodies might be present; if all reactions appear the directed against a high-frequency antigen. Using a case same, then there may be an autoantibody or an study approach, the first part of this review will cover antibody to a high-frequency antigen present. what steps can be used to detect and identify any A panel is performed and all RBCs are reactive; the alloantibodies underlying autoantibodies and how to autologous RBCs are nonreactive. The gel cards should differentiate multiple alloantibodies from antibodies be reviewed to determine if the reactions are equal. In directed against high-frequency antigens. If it is this case, the gel reactions are equal. The case is a determined that an alloantibody is directed against a referred to the local IRL and an anti-Yt is identified. In high-frequency antigen, then it becomes important to the interim period between sample submission and know the likelihood of that antibody being clinically resolution, the patient receives four units of significant, because antigen-negative RBCs may be incompatible blood. Over the next 3 weeks, the Hb difficult to obtain for transfusion. The second part of gradually falls and the patient receives two units of this review will focus on in vitro cellular methods used Yt(a–) blood. This review will focus on further investigation of to determine the clinical significance of alloantibodies. situations involving cases that present serologically It is 2 a.m. on the overnight shift and you are with all reagent RBCs reactive, including or not alone in the blood bank with a specimen drawn including the autologous RBCs. The starting point will from a patient in the emergency department who be in the hospital to highlight when, in each has a gunshot wound. This patient has a history circumstance, it would be advisable to send the sample of previous admissions and transfusions at your to the IRL. hospital. The antibody screen is 3+ with screening cells I, II, and III by the gel method. The Autoantibodies emergency department is notified of the positive The presence of an autoantibody is usually screen and gives an indication that blood is determined by a reactive autologous control in a needed for transfusion; the patient’s patient who has not been recently transfused. This is holding at 5g/dL but he is symptomatic. reactivity could be primarily present at room The antibody’s specificity should be identified;how temperature (probable cold autoantibody) or primarily this is done depends on the resources available at the present at 37°C and detected at the antihuman globulin

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 147

S.T.NANCE AND P.A.ARNDT

(AHG) phase of testing (probable warm autoantibody). directed at an enzyme-sensitive antigen) or adding LISS It is “best practice” to confirm that the reactivity is due or PEG to the serum-RBC mixture.5–11 The use of PEG to an autoantibody by performing an autoadsorption for adsorptions has been the topic of controversy and showing that the reactivity is removed; the because, in some studies, loss of alloantibody reactivity autoadsorbed sample can then be tested to determine has been reported.7,8,12 Adsorptions can be performed if alloantibodies are also present. Autoadsorptions are at 37°C or at 4°C to remove primarily warm or cold performed using autologous RBCs that have been autoantibodies. A combination of temperatures is often treated (e.g., with heat, enzyme, or ZZAP [a used for those antibodies showing reactivity in room combination of enzyme and DTT]) to remove some of temperature (RT) and 37°C phases (sometimes referred the globulin coating on the RBCs and/or to increase the to as warm/cold or combination autoantibodies). The capacity of the RBCs to adsorb autoantibody from the selection of the number of adsorptions that one serum. If the patient has been transfused recently,e.g., performs varies between IRLs; some do one more in the last 3 or 4 months, then autologous RBCs must adsorption than the strength of the serum reactivity be obtained to determine if the reactivity is (e.g.,two adsorptions for an antibody reacting 1+;some autoantibody prior to reporting it as such. Two use more adsorptions if testing is performed with methods used to obtain autologous RBCs from PEG), and some use the strength of the DAT to posttransfusion samples are reticulocyte separation by determine the number of adsorptions. Others monitor weight-based gradient separation1 and separation of the strength of the DAT on the adsorbing cells or test hemoglobin S positive RBCs by hypotonic wash.2 the adsorbed serum against fresh adsorbing cells to Once the autologous RBCs are obtained, they may be determine if more adsorptions need to be done. This insufficient in volume to use for routine adsorption but has the disadvantage of delaying adsorptions and they can be cleared of coating globulins and tested makes the turnaround time longer. with the patient’s serum and eluate to determine if there is reactivity directed against the patient’s RBCs. 37°C reactive autoantibodies If the patient has been transfused in the last 3 to 4 Case #1: A 68-year-old African American man is months, then an autoadsorption should not be admitted to the emergency room (ER) with a Hb of performed to determine if underlying alloantibodies 4.5 g/dL. He is pale and short of breath. The are present,because a small amount of transfused RBCs admitting resident requests four units of leuko- can adsorb alloantibody.3 One method that can be reduced RBCs as soon as possible. The antibody used to determine if alloantibodies are present in a screen is 3+ positive at the AHG phase with all three transfused patient with autoantibodies is allogeneic screening RBCs. Table 1 shows the initial panel adsorption (adsorption of the patient’s serum/plasma results, with reactivity with a few RBCs at the 37°C with allogeneic RBCs).4 The selection of RBCs for the phase, corresponding to anti-E specificity, and adsorptions is critical; there must be adsorption of reactivity with all RBCs at the AHG phase, including autoantibody onto RBCs expressing some antigens and the autologous control. The hospital refers the sample lacking others so that the commonly encountered, to the local blood center IRL. The ER later calls to say potentially clinically significant antibodies can be that the patient was transfused a month ago at identified in the adsorbed serum. The inherent risk another hospital. The hospital blood bank calls the with allogeneic adsorptions is that, if there is an technologist at the IRL, who is just finishing the antibody to a high-frequency antigen, it will be testing on the autoadsorbed serum. Anti-E is present adsorbed by the allogeneic RBCs and not be in LISS at 37°C and AHG phases (see results in Table detectable. Usually, the following RBC samples are 2). Because the patient has been recently transfused, chosen: R1,R2, and rr; at least one sample lacking the IRL repeats the adsorptions using enzyme-treated K; randomly selected RBCs negative for Jka,Jkb,S,s; allogeneic RBCs. Four allogeneic adsorptions are and RBCs treated with enzyme or ZZAP so that they performed using the RBC samples listed in the bottom are effectively negative for Fya,Fyb,M,and N. In general, of Table 3. The results of the allogeneic adsorptions a b consideration is usually not given to Le ,Le, or P1 are shown in Table 3. The results show that the unless there is a suspicion that antibodies to those patient has an antibody reactive with all RBCs tested antigens are present. Variations of allogeneic adsorptions (from the initial panel, Table 1), the suspected anti-E, include utilizing untreated cells (if the autoantibody is and something else, possibly anti-K (reactive against

148 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 Review: all RBCs incompatible—serologic aspects

Table 1. Initial panel results—case #1 a b a b a b Panel cells D C E c e f K k Fy Fy Jk Jk MN S sLuLu P1 IS 37° LISS Anti-IgG 1 ++00+00+0++++0++0++ 0 0 3+ 2 +00++++0++0+0+0+0++ 0 0 3+ 3 +0++000++0++++++0+0 0 2+3+ 4 0+0+++0+0++00++00++ 0 0 3+ 5 00++++0+0+0+++0+0++ 0 2+3+ 6 +00+++0+0+0+++0+0+0 0 0 3+ 7 +0++++0+++0++++00++ 0 2+3+ 8 000++++++++0+++00++ 0 0 3+ 9 000+++++0++0++++0++ 0 0 3+ 10 000+++0++0+++00+0++ 0 0 3+ 11 000+++0+00+++00+0++ 0 0 3+ Auto 00 4+

Table 2. IRL testing on the autoadsorbed serum—case #1

a b a b Panel cells D C E c e f K k Fy Fy Jk Jk MN S sP1 IS 37°C LISS Anti-IgG 1 +0++++0+0++00+++0 0 2+ 2+ 2 ++00+0+0+00++++++ 0 0 0 3 + + 0 0 + 0 0 + 0 +0++0++0 0 0 0 4 + 0 + + 00++0++0+0+++ 0 2+ 2+ 5 ++0+++0+++++0+0++ 0 0 0 6 0+0+++0++0+++++00 0 0 0 7 00++++0+0++0++0++ 0 2+ 2+

Table 3. The results of four allogeneic adsorptions—case #1 Serum adsorbed with rr R1 R2

LISS LISS LISS a b a b Panel cells D C E c e f K k Fy Fy Jk Jk MN S s P1 37°C 37°C 37°C Anti-IgG Anti-IgG Anti-IgG 1 +0++++0+0++00+++ 0 02+0 0 02+ 2 ++00+0+0+00+++++ + 01+01+01+ 3 ++00+00+0+0++0++ 0 0 0 0 0 0 0 4 +0++00++0++0+0++ + 02+0 0 02+ 5 ++0+++0+++++0+0+ + 0 0 0 0 0 0 6 0+0+++0++0+++++0 0 0 0 0 0 0 0 7 00++++0+0++0++0+ + 02+0 0 02+

R1* 00000

R2*000 0 0 rr* 0 0 0000 *enzyme-treated RBCs used for the allogenic adsorptions

RBC suspension #2). Another possibility is that this autoantibody even though autoadsorption was not reactivity is due to an HLA antibody. Further tests valid due to the presence of transfused RBCs. show that this is an anti-K. The anti-K was not Case Comments: Results in this case suggest that present in the autoadsorbed serum, perhaps due to the patient’s broadly reactive antibody has auto- remaining transfused RBCs in the patient’s sample specificity; the proof of an autoantibody would be adsorbing the weak reactivity. The reactivity with adsorption of the antibody onto autologous RBCs. In all RBCs tested (Table 1) is presumed to be due to an the case of a transfused patient, autologous cell

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 149

S.T.NANCE AND P.A.ARNDT separations would need to be done to isolate the temperatures (4°C) is the method of choice to resolve autologous RBCs. Tests with autologous RBCs and the cases of suspected cold autoantibodies. If the patient patient’s serum or eluate are expected to be positive if has been transfused, allogeneic adsorption in cold an autoantibody is present. If they are negative, then it temperatures as well as use of rabbit RBCs for should be determined if an antibody to an antigen of adsorption can be used. It is essential to read package high frequency is present. The allogeneic adsorptions inserts for use of rabbit RBCs or stroma, as other would have adsorbed an antibody to a high-frequency antibodies may also be adsorbed. antigen. If the unexplained reactivity (with RBC Case #2: A sample is received in the blood bank suspension #2) of the adsorbed serum had not been from a patient scheduled for cardiac surgery the next shown to be anti-K,then tests with chloroquine-treated day. The antibody screen by gel shows strong RBCs would have been performed to determine if the reactivity with many RBCs in the top layers of the gel reactivity was due to HLA antibodies. and some RBCs streaming through the gel in all tests. The panel gives similar results (all RBCs are reactive) Cold-reactive autoantibodies and the sample is sent to the local IRL, as the hospital Some laboratories approach the detection of does not test in tubes and has exhausted its resources. underlying alloantibodies, in the presence of suspected The IRL’s routine approach involves tube testing in cold-reactive antibodies, by using a prewarming albumin (immediate spin [IS], RT for 15 minutes, method. This is not recommended until it has been 37°C for 30 minutes with albumin, and AHG phase proved by autoadsorption that the antibody is an using anti-IgG) and against ficin-treated cells (37°C autoantibody, as incorrect use of the technique has for 15 minutes, AHG phase with anti-IgG). The IRL been reported to yield incorrect results.13 Excellent observes 1+ reactivity at IS, 3+ at RT, 1+ discussions of the pros and cons of prewarming were agglutination and a trace of hemolysis at 37°C, and published in 1995 and 1996.14–16 Once the auto- +w reactivity at the AHG phase that could reflect reactivity of the broadly reactive antibody is defined, carryover from 37ºC (see Table 4). The ficin-treated prewarmed testing may be of value in the assessment RBC panel was completely hemolyzed at 37°C and no of underlying alloantibodies and in the characterization RBCs were left to read at the AHG phase. The DAT was of the clinical significance of the reactivity. Caution is 3+ with anti-C3. Four adsorptions with autologous advised, however, as the loss of potentially clinically RBCs treated with ZZAP were performed at 4°C. In significant alloantibody reactivity in prewarmed testing this case, the patient’s sample was sufficient to allow has been described.17–19 Autoadsorption at cold four aliquots of treated autologous RBCs to be

Table 4. IRL results of panels using serum from case #2 Albumin Ficin

a b a b Panel cells D C E c e f K k Fy Fy Jk Jk MN S s P1 IS RT 37°C 37°C Anti-IgG Anti-IgG 1 + + 0 0 + 0 0 + 0 + + + + 0 + + + 1+ 3+ 1+th* +w h† nc‡ 2 +00++++0++0+0+0+ +1+3+1+th+whnc 3 + 0 + + 0 0 0 + + 0 + + + + + + 0 1+ 3+ 1+th +w h nc 4 0 + 0 + + + 0 + 0 + + 0 0 + + 0 + 1+ 3+ 1+th +w h nc 5 0 0 + + + + 0 + 0 + 0 + + + 0 + + 1+ 3+ 1+th +w h nc 6 +00+++0+0+0+++0+ 01+3+1+th+whnc 7 + 0 + + + + 0 + + + 0 + + + + 0 + 1+ 3+ 1+th +w h nc 8 0 0 0 + + + + + + + + 0 + + + 0 + 1+ 3+ 1+th +w h nc 9 0 0 + + + + + + 0 + + 0 + + + + + 1+ 3+ 1+th +w h nc 10 0 0 0 + + + 0 + + 0 + + + 0 0 + + 1+ 3+ 1+th +w h nc 11 0 0 0 + + + 0 + 0 0 + + + 0 0 + + 1+ 3+ 1+th +w h nc Auto 1+ 3+ 1+th +w h nc *Trace hemolysis †Completely hemolyzed ‡No cells remaining

150 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

Review: all RBCs incompatible—serologic aspects prepared. In some cases, insufficient RBCs are concern for cardiac surgery than for other surgeries available, due either to the patient’s Hb or to the because the temperature of the patient is lowered in sample size submitted. In those cases, it may be cardiac surgery.20 necessary to use the same aliquot of autologous RBCs over and over and re-treat the aliquot between Multiple Antibody Specificities adsorptions. The autoadsorbed serum was Often the presence of multiple antibodies can be nonreactive when tested with a set of three antibody discerned by the differences seen in serologic phases screening cells. When the report of a cold or strengths of reactivity with panel RBCs in tube autoantibody was given to the hospital, the cardiac testing and in the visual appearance of positive surgeon requested a cold agglutinin titer and reactions in gel testing. Depending upon the thermal amplitude study to determine if this would configuration of the panel and the antibodies present interfere with the surgery and the plans to use cell in the patient’s sample, nonreactive cells may be salvage techniques. A thermal amplitude study was obtained or reactivity indicating dosage effects or performed with dilutions of the patient’s serum in separate antibodies may be discerned. These features saline with the following results (titers): 30°C = 8, can be used to differentiate the presence of multiple 22°C = 64, 4°C > 2048. The surgery could not be antibody specificities from an antibody to a high- postponed. After consultation with the medical frequency antigen. The use of enzyme-treated RBCs director of the IRL, hospital blood bank physician, can also be helpful in differentiating antibody and surgeon, it was decided that the apparent cold specificities. agglutinin might be a problem with cell salvage at RT Case #3: A sample from a patient with sickle cell and cold cardioplegia might result in agglutinated disease was received with a request for two units of autologous RBCs in the coronary arteries. In the blood for outpatient transfusion the next day. The surgery, the blood was first drained from the heart antibody screen was positive. Cell I was 2+, cell II and then cold was applied. Cell salvage was not used was 1+, and cell III was 3+ in the AHG phase. A panel and the temperature was not lowered below normal was tested and there was variable reactivity with 16 RT. The patient did well and received two units of of 16 RBC suspensions tested. The blood bank blood, which were given through a blood warmer. requested that the IRL evaluate the sample, as it was Case Comments: This patient had serologic apparent the hospital would not have sufficient RBC results consistent with a diagnosis of cold agglutinin panels to evaluate the sample, even though they syndrome; the patient’s Hct was 33%. This had not saved the most recently expired panels. At the IRL, the been detected prior to scheduling surgery. The patient’s RBCs typed as D+C–E–c+e+ in the routine patient’s pathologic cold agglutinin was more of a Rh phenotype that is performed on all new patients.

Table 5. IRL special panel configured for case #3 LISS Ficin

a b a b a b Panel cells D C E c e f K k Fy Fy Jk Jk MN S sLuLu P1 IS 37°C 37°C Anti-IgG Anti-IgG 1+00+++0 ++00+++++0++ 0 0 2+0 0√ 2+00+++0 ++00+++0+0+0 0 0 2+0 0√ 3000+++0 +0++0++0+0++ 0 0 1+0 3+ 4 0 0 0 + + + 0 + 0 + +00+++0++ 0 0 1+0 3+ 5000+++0 +000++0+00++ 0 0 0√ 00√ 6+00+++0 +000+0+0+0+0 0 0 0√ 00√ 7+00++++ +000++0++0++ 0 1+3+2+3+ 8000++++ +0+0++++00++ 0 1+3+2+3+ 9 + + 0 0 + 0 0 + 0 + 0 +++++0++ 0 1+2+2+3+ 10 + + 0 0 +00 + 0+0+++++0+0 0 1+2+2+3+ 11 + 0++000 +0+0+++0+0++ 0 0 1+2+4+ 12 + 0++000 +0+0+++0+0++ 0 0 1+2+4+

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 151 S.T.NANCE AND P.A.ARNDT

The IRL panel was configured by the staff so that the adsorptions does increase the risk of diluting the serum first six RBC suspensions were C–E–K– and the rest of and thus missing weak antibodies. the panel (six RBC suspensions) was designed to rule in anti-C, -E, or -K if they were present. The routine Antibodies to High-Frequency Antigens testing at the IRL included a LISS panel and a ficin- Case #4: A 59-year-old Caucasian man was on treated RBC panel run in parallel. The results are dialysis for kidney failure. Two units of blood were shown in Table 5. The use of enzyme-treated RBCs requested for transfusion with his next dialysis a demonstrated the likelihood of anti-Fy . Subsequent procedure (EPO was not yet available). The antibody a a studies proved that anti-C, -E, -K, -Fy , and -Jk were screen was positive (3+ with all cells by the IAT). A present in this patient’s sample. panel was tested and all RBCs were reactive (3+); Case Comments: Utilizing information obtained autologous RBCs were nonreactive. Samples were from the patient’s Rh phenotype made antibody sent to the IRL. identification easier. Additionally, the IRL studied the Case #5: A 36-year-old Hispanic woman had a panel that had been performed at the hospital and saw history of six pregnancies and four transfusions that there was variation in reactivity. With the (associated with two of the deliveries). There was no knowledge that the first antibodies formed often have history of HDN. She was admitted with a diagnosis Rh specificity,that the K antigen is very immunogenic, of spontaneous abortion. The antibody screen and and that the hospital panel indicated those specificities panel were positive (2+ with all cells by IAT); could cause the reactivities seen, a custom panel was autologous RBCs were nonreactive. Samples were designed. Since the IRL obtains panels from all sent to the IRL. manufacturers, it was easy to make a “custom”panel to When antibodies to antigens of high frequency are perform the testing. Custom panel configuration is present, the resolution can be time- and resource- facilitated with computer software called Antigen Plus consuming. The first sign that an antibody to a high- Ab-ID (Rowny Systems, Inc., Gaithersburg, MD), which frequency antigen is present is usually uniform allows the user to select the RBCs (by phenotype) for reactivity with all reagent RBCs tested; commonly, this testing. This program utilizes electronic information reactivity is only at the antiglobulin phase of testing. In from the panel manufacturer (Immucor/Gamma, the majority of cases, the autologous RBCs are Norcross, GA; Olympus America, Melville, NY), which nonreactive,but sometimes they are reactive due to the allows mistake-free entry into Antigen Plus. Ortho- presence of recently transfused RBCs (the reaction may Clinical Diagnostics (Raritan, NJ) does not yet provide have a mixed-field appearance) or due to another this information; the user must manually enter the unrelated cause (medication, nonspecific, etc.). phenotype information from their panels. Then the Approaches to resolution of the antibody’s specificity user requests the desired phenotype of RBCs and the include testing RBCs that have been treated with software peruses the database and presents choices to different agents, e.g., enzyme or DTT, and looking for the user. In this way, only RBCs that are of the needed enhancement or diminution of reactivity. Additionally, phenotype are tested, thus conserving serum (and the even though the use of a high titer value to steer the reagent RBCs); the phenotype information on the investigation toward a Knops (KN) system antibody is selected RBCs is made into a panel worksheet by the downplayed by some experts,one of the authors (STN) software, thus preventing handwriting and associated still finds it valuable. If the antibody’s titer is equal to errors. The information is presented in a panel format or greater than 32, DTT-treated RBCs are nonreactive, with space for recording test results. and the KN null cell is nonreactive, STN’s laboratory By using all the resources available, the IRL was reports the antibody as a probable KN system antibody able to resolve the case by testing two panels in about and stops the investigation. This approach was 3 hours. If the IRL does not have access to RBCs with approved by the Technical Advisory Committee the phenotypes needed for testing, allogeneic (customers). adsorptions of the patient’s serum may be useful to In the evaluation of an antibody to a high- adsorb out some specificities and leave others behind. frequency antigen, the IRL’s frozen rare RBC and serum It should be remembered that very strongly reactive resources are usually utilized to the full extent to antibodies may require high numbers of adsorptions to identify the antibody. If the IRL does not have ABO- be completely removed. Increasing the number of compatible RBCs negative for all high-frequency

152 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 Review: all RBCs incompatible—serologic aspects antigens for testing against the patient’s serum, then from the first aliquot of adsorbing RBCs can be used to the presence or absence of those antigens on the test for the antibody to the high-frequency antigen, patient’s RBCs becomes important. It is helpful to once any antibodies to common antigens are identified. know the patient’s race when choosing which antigens The investigation may be assisted through the use to test for and it is important to know the patient’s of different media (e.g., ). Frequently, the use transfusion history when interpreting the RBC typing of a combination of phenotyping the patient’s RBCs results. Although IRLs are continually seeking to freeze (e.g., for Jsb, S, s, Kpb, I, Jka,Jkb,Lub, H) and testing the more rare sera and RBCs, these are generally not patient’s serum against “null” RBCs (e.g., cord, Oh,or available commercially and are obtained through DTT-treated [Ko] RBCs) is valuable. Knowledge of the resource sharing groups like SCARF and an exchange race of the patient may be helpful early in the program in the American Red Cross IRLs known as investigation to choose the more common RBCs TRANSFERASE. Some of the “more common” high- negative for high-frequency antigens known to exist in frequency, antigen-negative RBCs that may be available that ethnic group (e.g., U–, Sl[a–], Js[b–] RBCs for on commercial panels are k–, I–, Yt(a–), Js(b–), and African Americans). Other clues include reactivity at k Kp(b–), especially if a review of recently outdated RT (e.g., anti-H, -PP1P ) or hemolysis of RBCs (e.g., anti- panels is done. If expired panel RBCs are used and a Vel, -P). See Table 6 for a list of antibodies to high- negative reaction is obtained, then additional testing frequency antigens that are suggested by some of the using in-date RBCs or frozen/thawed RBCs is indicated. clues mentioned above. This is not a complete list and If there is a suspicion that a sample contains an the reader is referred to the Blood Group Antigen antibody to a high-frequency antigen and underlying FactsBook.21 If such investigation is unproductive, alloantibodies, it is advisable to identify the antibodies testing of the entire collection of RBCs negative for to common antigens first. Test results of RBCs negative high-frequency antigens or molecular studies are often for high-frequency antigens may be unproductive if the the solution. RBCs are positive for a common antigen that the Once the specificity of an antibody to a high- patient also has an antibody against. Underlying frequency antigen is identified, then the clinical alloantibodies can be identified by first phenotyping significance must be determined so the hospital can the patient for common antigens, then selecting a know what to transfuse. There are excellent texts that phenotype-similar RBC sample for adsorptions. If review the literature on this.21–23 For antibodies that are untreated RBCs are used for the first adsorption, then known not to be clinically significant, by either an eluate can be prepared from the adsorbing RBCs, specificity or phase of reactivity, transfusion of random and a pure source of the antibody to the high- RBCs is recommended. After transfusion, serologic frequency antigen will be available. The adsorbed reevaluation of the patient’s sample should be serum should be tested back against the adsorbing performed for exclusion of other newly developed RBCs for evidence of complete adsorption, as alloantibodies. Re-identification of existing antibodies alloantibodies are more variable in their strength (in is optional. comparison to most autoantibodies, which have If the antibody is known to be clinically significant relatively low titers). The adsorbed serum can be used and blood availability is usually not a problem (e.g., k– for identification of antibodies against common or Kp[b–] RBCs), then a request can be made to the antigens that the patient’s RBCs lack, and the eluate local blood supplier and blood can be obtained. If the

Table 6. Some antibodies to high-frequency antigens to consider Room Decreased African Asian, Hispanic, temperature Hemolysis reactivity in American Caucasian Native American reactivity seen enzyme patient patient patient Anti-H Anti-Vel Anti-Ch Anti-U Anti-Kpb Anti-Dib

k b a Anti-I Anti-PP1P Anti-Rg Anti-Js Anti-Yt k b a Anti-PP1P Anti-P Anti-In Anti-Jo Anti-Vel Anti-Jk3 Anti-JMH Anti-Hy Anti-Sda Anti-H Anti-EnaTS Anti-Cra Anti-P Anti-I Anti-EnaFS Anti-Tca Anti-Ata

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 153 S.T.NANCE AND P.A.ARNDT blood is not immediately available from the local blood specificity and thermal range), IgG subclassing supplier,the request can go to an American Rare Donor methods, in vivo survival studies (e.g., 51Cr and the Program member facility. The reader is referred to the “biological crossmatch”), and in vitro cellular assays. next issue of Immunohematology for a discussion of This review will concentrate only on the latter, the in the American Rare Donor Program and the ISBT Rare vitro cellular assays. These include the antibody- Donor Program. dependent cellular (ADCC) assay33 (which If the antibody is directed against an antigen of has only been used to assess the clinical significance of variable clinical significance, and/or the blood is not antibodies in the setting of HDFN), the chemi- immediately available, then in vitro assays to predict in luminescence test (CLT),34 and the monocyte mono- vivo clinical significance should be used. These studies layer assay (MMA)32,35–37; most of this review will focus are valuable; if results are negative, blood may be on the MMA. available as liquid random units, thus reducing the The premise of all in vitro cellular assays is to mimic logistics and time needed for transfusion, and the rare in the laboratory what occurs in vivo when antigen- units can be saved for those patients who cannot positive RBCs are transfused into a patient with the receive random units. Patients who should not receive corresponding antibody. If the antibody-coated RBCs random units are those who have had either a positive are destroyed, it will be by one of two mechanisms: in vitro test, a transfusion reaction to random units, or intravascular or extravascular.20 Intra-vascular RBC both. destruction (hemolysis of RBCs directly in the bloodstream) is a rare event due to the action of Methods Used to Determine the Clinical complement on the RBC membrane.20 Most antibodies Significance of Alloantibodies do not cause RBC destruction via this mechanism, but Before describing methods used to determine the rather by the extravascular mechanism, which involves clinical significance of alloantibodies, it is important to destruction of RBCs via in the liver or discuss what is meant by the term “clinically . Macrophages recognize and attach to RBCs that significant” in relation to alloantibodies and RBCs.24,25 are coated with IgG and/or C3 via specific receptors Everyone would agree that an antibody that causes an on the .20 The attached RBCs are then obvious, clinical hemolytic transfusion reaction (fever, destroyed in three ways: the RBCs can be completely chills, hemoglobinemia, hemoglobinuria, etc.) is a engulfed (phagocytized) by the macro-phage, the RBCs clinically significant antibody and that antigen-negative can be partially phagocytized and the resulting RBCs are needed for transfusion. What about an spherocytes trapped and destroyed in the spleen,or the antibody that does not cause any overt clinical RBCs can be lysed extracellularly by enzymes secreted symptoms, but is associated only with laboratory signs by the macrophage.20 In vitro cellular assays attempt of hemolysis (increased bilirubin, decreased to mimic extravascular RBC destruction. , etc.)? Or an antibody that is not In all in vitro cellular assays, RBCs are sensitized associated with any clinical or laboratory signs of with the antibody in question and are then incubated hemolysis, but RBCs incompatible with it survive less with mononuclear cells (usually monocytes, the than their normal lifespan? The definition of “clinical precursors of macrophages). The in vitro reactions of significance” may vary depending upon the needs of the monocytes with the antibody-coated RBCs are then the patient. For a patient with a short-term ascertained by (1) visual inspection for attachment requirement for blood, e.g., a surgical patient, it may and/or of RBCs by monocytes (MMA), (2) not be important for the transfused RBCs to have measurement of oxygen radicals released by mono- completely normal survival. On the other hand, for a cytes during phagocytosis (CLT), or (3) measurement patient with a hematological disorder and long-term of direct hemolysis of 51Cr-labeled RBCs (ADCC). The requirements for blood, it would be more important MMA and CLT assays, which have been used to that the blood survive as long as possible, to limit the determine clinical significance of alloantibodies for total number of transfusions. transfusion purposes, have a cutpoint, or value above The methods used to determine the clinical which the test is considered to be positive. When a significance of RBC alloantibodies have been reviewed positive result is obtained, it is recommended that elsewhere.24,26–32 They include routine serologic antigen-negative blood be transfused in order to avoid methods (which give information about an antibody’s an overt transfusion reaction.

154 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

Review: all RBCs incompatible—serologic aspects

A number of variations of the MMA have been MMAs becoming positive after transfusion,43,44 as well performed by different investigators over the past 25+ as of positive MMAs becoming negative after years.35–37 Many of the variables involved in the MMA transfusion.45,46 In general, once a positive MMA result have been reviewed38; they include the source of is obtained, further MMAs are not performed. monocytes,36,39 the RBC sensitization procedure Case #4 (continued): The antibody detected in (including whether or not fresh normal serum as a the Caucasian patient’s serum was identified as anti- source of complement is added40,41), the “culture” Ge.The patient received Ge– units for 10 years, until conditions (e.g., a CO2 atmosphere versus ambient the supply of Ge– RBCs in the country was almost air42), and how the results are expressed. The assay depleted. An MMA was requested and found to be currently used by the American Red Cross (Southern negative (2.8%). The patient received two Ge+ units California and Penn-Jersey Regions) is as follows: during dialysis with no untoward reaction noted. mononuclear cells from one or more donors are The Hb rose as expected and there was no rise in isolated using Ficoll®-Hypaque and incubated in bilirubin. One week after the transfusion, the MMA ambient air on a glass slide, the nonadherant was repeated and was still negative (2.7%); the lymphocytes are suctioned off, the sensitized RBCs antibody reactivity was 2+. The patient received two (sensitized with and without fresh complement more Ge+ units without an obvious reaction and present) are added to the monocyte monolayer on the there was an expected rise in Hb. The MMA was slide and incubated in ambient air; the nonadherant repeated three more times over the next several RBCs are removed (via suctioning and washing), and months. Each time the MMA was negative and the the slide is stained and examined microscopically for patient was able to successfully receive Ge+ RBCs. the percentage of monocytes with RBCs adhering The strength of the anti-Ge weakened; it was only and/or phagocytized.37 microscopically reactive in the last sample. The When the assay currently used by the American patient was then started on EPO and required no Red Cross was originally set up,a cutpoint of 3 percent further transfusions. monocyte reactivity was selected based on the results Case #5 (continued): The antibody in the of unsensitized RBCs in the assay.37 The Southern Hispanic woman’s serum was identified as anti-Yta. California Region later used statistical methods and An MMA was requested and found to be negative data from patients who had received incompatible (0%). Two months later, during an emergency, the transfusions to determine that a more appropriate patient was transfused with two Yt(a+) units of cutpoint for their assay was 5 percent reactivity.25 blood. Her DAT became transiently positive but no Using this 5 percent cutpoint, the sensitivity of the clinical symptoms of a reaction were observed. A MMA is 100 percent (no false negatives) and the month later another MMA was requested in specificity is 61 percent (there are false positives).25 anticipation of a scheduled hysterectomy. The MMA If a result greater than the cutpoint is obtained was now positive (23%) and the patient received two when testing an antibody,the MMA is considered to be Yt(a–) units during her surgery. positive, and to avoid an overt transfusion reaction, These two patients demonstrated different antigen-negative blood is recommended as the optimal outcomes after transfusion of incompatible RBCs. choice for transfusion. When a result less than the Patient #4’s antibody became progressively weaker cutpoint is obtained when testing an antibody, the and the MMAs remained negative. Patient #5’s MMA is considered to be negative;that antibody should antibody changed its characteristics so that antigen- not cause acute hemolysis of transfused antigen- negative blood became the better choice for positive RBCs at that time. A negative MMA, however, transfusion. does not guarantee that transfused,sensitized RBCs will Tables 7 and 8 summarize data from published have normal long-term survival. If a patient with a reports on alloantibodies to high-frequency antigens negative MMA is transfused with antigen-positive RBCs, that may be of variable or unknown clinical the exposure to more antigen may cause the antibody significance. In Table 7, MMA results from 29 such to change its characteristics (e.g., strength/titer or antibodies are compared with the results of 51Cr subclass composition). Thus, the MMA must be survival studies. 51Cr survival study results are repeated in these patients prior to transfusion of more commonly expressed as the percentage of antigen-positive RBCs. There are reports of negative incompatible RBCs surviving at 1 hour and 24 hours

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 155 S.T.NANCE AND P.A.ARNDT

Table 7. Data from reports in the literature comparing MMA results of antibodies to high-frequency completely compatible red cells, antigens with 51Cr survival studies. Results for the 16 antibodies with 1-hour 51Cr survival study results available are listed under the “1-hour 51Cr” heading (shaded columns) as either donor red cells may be transfused normal (N) or abnormal (Ab). The results for the remaining 13 antibodies without 1-hour 51Cr with minimal hazard when, 51 results are listed under the “ Cr survival” heading. following a test with 0.5 ml of the Antibody MMA = Negative MMA = Positive Specificity 1-hour 51Cr 51Cr survival 1-hour 51Cr 51Cr survival donor’s red cells, . . . the red-cell (references) # N Ab N Ab N Ab N Ab survival at 60 minutes is not less Yta 25,35,37,43,47 910203102than 70%.” It is known that a 51 Ge36,37,44,48,49 511020001normal 1-hour Cr survival result Lub 50 100010000does not mean that the transfused Lu651–53 300101010incompatible RBCs will survive Lu1254 110000000normally (i.e.,the 24-hour survival Cra 55,56 200001001or T50Cr may be abnormal). As it 51 Tca 45 110000000can be difficult to have Cr Tcab 57 100000001survival studies performed, in vitro assays (e.g., the MMA) were Lan37,58 200000200 developed to obtain the same Ata 59,60 200001100 information as a 1-hour 51Cr Hya 61 110000000 survival.24 Of the 29 reports in Oka 62 100000001 Table 7, only 16 included the 1- TOTAL2951336416hour 51Cr survival data. Results in Table 7 are divided into those after transfusion of a small dose of RBCs and/or as the which had 1-hour 51Cr survival data available and those half-life of those RBCs (T50Cr). The International that did not (e.g., data were available from other times Committee for Standardization in Haematology76 posttransfusion or the authors summarized the published the following guideline with respect to using interpretation of the 51Cr survival study without giving the 51Cr survival study as a compatibility test: “In cases actual data). of urgency or when there is great difficulty in finding Although most MMA and 51Cr results concurred, there are eight results in Table 7 that appear Table 8. Data from reports in the literature comparing MMA results with result of transfusion (txn) of incompatible RBCs. Results in the “Hemolytic txn, MMA to be discrepant:one antibody with a negative positive reaction” column have been further subdivided into those described MMA had an abnormal 1-hour 51Cr survival as having (immediate or clinical signs/mild, delayed or laboratory signs only/no information given). study and seven antibodies with positive 51 Antibody MMA = Negative MMA = Positive MMAs had normal Cr survival results (six Specificity No txn Hemolytic No txn Hemolytic had normal 1-hour results). The one antibody (references) N reaction txn reaction reaction txn reaction with the negative MMA and abnormal 1-hour Yta 24,34,35,37,38,47,63,64 31 22 0 5 4 (1/3/0) 51Cr result was an anti-Ge in an untransfused Ge34,36,37,65,66 6 5 0 0 1 (0/0/1) male48; the MMA was negative with native Cra 67,68 4 2 0 1 1 (0/1/0) serum but was positive when concentrated Tca 69 1 0 0 0 1 (0/1/0) serum was used for testing. The six Lub 46 1 0 0 0 1 (0/1/0) antibodies (three anti-Yta and one each anti- Lu651 11000Lu6, -Cra, and -Ata) with positive MMAs and Lu870 1 0 0 0 1(1/0/0) normal 1-hour 51Cr survival results had Hy35 1 0 0 0 1 (0/0/1) abnormal 51Cr results after the 1-hour result. Gy50 1 0 0 0 1 (0/1/0) The other antibody (anti-Lu6) with a positive 51 Joa 22 1 0 010MMA and a normal Cr result was associated 53 Jra 35,71,72 4 3 0 0 1 (1/0/0) with a median survival of 26 days, and thus Lan37 11000appears to be falsely positive. Dib 73 1 0 0 0 1 (0/1/0) Table 8 shows data on 56 antibodies Dra 74 1 1 0 0 0 where MMAs were performed and the Era 75 11000patients were transfused with incompatible TOTAL 56 35 0 7 13 (3/8/2) RBCs (unfortunately, in many cases it is not

156 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

Review: all RBCs incompatible—serologic aspects known if the MMA was performed before or after the 4. Allogeneic red cell adsorption for removal of transfusion). In these cases, information was given as warm autoantibody. In: Immunohematology to whether or not there was evidence for a hemolytic methods and procedures. 1st ed. The American transfusion reaction, but often not many details were National Red Cross 1993:47-51. given. The results in Table 8 listed under the 5. Liew YW, Duncan N. Polyethylene glycol in auto- “Hemolytic transfusion reaction” heading are adsorption of serum for the detection of subdivided into those said to have an immediate alloantibodies (letter).Transfusion 1995;35:713. reaction or clinical signs; those with a mild, delayed 6. Chiaroni J,Touinssi M, Mazet M, deMicco P,Ferrera reaction or only laboratory signs of a reaction; and V. Adsorption of autoantibodies in the presence of LISS to detect alloantibodies underlying warm those with no detailed information. Although most autoantibodies. Transfusion 2003;43:651-5. MMA results agreed with the results of transfusion, 7. Barron CL, Brown MB. The use of polyethylene there appear to be seven discrepant results in Table 8: glycol (PEG) to enhance the adsorption of auto- seven patients with positive MMAs had no signs of a antibodies. Immunohematology 1997;13:119-22. transfusion reaction and the MMA results appear to be 8. Judd WJ,Dake L. PEG adsorption of autoantibodies falsely positive. causes loss of concomitant alloantibody. There are other single-case reports in the literature, Immunohematology 2001;17:82-5. on patients with antibodies to high-frequency antigens 9. Leger RM, Garratty G. Evaluation of methods for with variable or unknown significance, where MMAs detecting alloantibodies underlying warm were performed but the patients were not transfused autoanti-bodies. Transfusion 1999;39:11-6. with incompatible blood. One study reported MMA 10. Branch DR, Petz LD. Detecting alloantibodies in data on 251 alloantibodies; 192 of these alloantibodies patients with autoantibodies (editorial). were directed against high-frequency antigens of Transfusion 1999;39:6-10. variable or unknown clinical significance.25 The 11. Cheng C-K,Wong ML, Lee AW. PEG adsorption of majority (68%) of these 192 alloantibodies were autoantibodies and detection of alloantibodies in positive in the MMA. Thus, one should presume that warm autoimmune hemolytic anemia. Transfusion most alloantibodies to high-frequency antigens have 2001;41:13-7. the potential to be clinically significant, unless proven 12. Leger RM,Ciesielski D,Garratty G.Effect of storage otherwise by MMA or 51Cr survival studies. on antibody reactivity after adsorption in the presence of polyethylene glycol (letter). Transfusion 1999;39:1272-3. Acknowledgments 13. Storry JR, Mallory D. Misidentification of anti-Vel We thank Margaret Manigly for her preparation of due to inappropriate use of prewarming and this manuscript. The authors would also like to express adsorption techniques. Immunohematology 1994; their sincere appreciation and admiration for George 10:83-6. Garratty, PhD, who steadfastly and energetically 14. Judd WJ. Controversies in transfusion medicine. mentored and supported us throughout our careers! Prewarmed techniques:Con. Transfusion 1995;35: 271-5. References 15. Mallory D. Controversies in transfusion medicine. Prewarmed tests: Pro—why, when, and how—not 1. Reid ME,Toy PT.Simplified method for recovery of if. Transfusion 1995;35:268-70. autologous red blood cells from transfused 16. Garratty G. Attack on prewarmed tests—Too patients. Am J Clin Pathol 1983;79:364-6. much hot air (letter)? Transfusion 1996;36:192. 2. Brown DJ. A rapid method for harvesting 17. Judd WJ, Steiner EA, Knaff P, Davenport RD. autologous red cells from patients with hemo- Failure to detect potentially significant antibodies globin S disease. Transfusion 1988;28:21-3. in prewarmed tests (abstract). Transfusion 3. Laine EP, Leger RM,Arndt PA, et al. In vitro studies 1995;35: (Suppl):68S. on the impact of transfusion on the detection of 18. Kilsby MA.Comparison of prewarmed techniques: alloantibodies after autoadsorption. Transfusion with and without enhancements (abstract). 2000;40:1384-7. Transfusion 1999;39(Suppl):104S.

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 157 S.T.NANCE AND P.A.ARNDT

19. Leger RM, Garratty G. Weakening or loss of reactions: a technical workshop. Washington DC: antibody reactivity after prewarm technique. American Association of Blood Banks, 1982: Transfusion 2003;43:1611-4. 91-119. 20. Petz LD, Garratty G. Immune hemolytic anemias. 33. Engelfriet CP, Ouwehand WH. ADCC and other 2nd ed. Philadelphia: Churchill Livingstone, 2004: cellular bioassays for predicting the clinical 133-65,352-6. significance of red cell alloantibodies. Baillieres 21. Reid M, Lomas-Francis C. The blood group antigen Clin Haematol 1990;3:321-37. factsbook. 2nd ed. San Diego: Elsevier Academic 34. Hadley A, Wilkes A, Poole J, Arndt P, Garratty G. Press, 2004. A chemiluminescence test for predicting the 22. Issitt PD,Anstee DJ. Applied blood group serology. outcome of transfusing incompatible blood. 4th ed. Durham: Montgomery 1998. Transfus Med 1999;9:337-42. 23. Daniels G. Human blood groups. 2nd ed. Oxford: 35.Schanfield MS, Stevens JO, Bauman D. The Blackwell Science, 2002. detection of clinically significant erythrocyte 24. Garratty G. Evaluating the clinical significance of alloantibodies using a human mononuclear blood group alloantibodies that are causing assay.Transfusion 1981;21:571-6. problems in pretransfusion testing. Vox Sang 36. Branch DR, Gallagher MT, Mison AP, Sy Siok Hian 1998; 74:285-90. AL, Petz LD. In vitro determination of red cell 25. Arndt PA, Garratty G. A retrospective analysis of alloantibody significance using an assay of the value of monocyte monolayer assay results for monocyte-macrophage interaction with sensitized predicting the clinical significance of blood group erythrocytes. Br J Haematol 1984;56:19-29. alloantibodies. Transfusion 2004 (in press). 37. Nance SJ, Arndt P, Garratty G. Predicting the 26. Daniels G, Poole J, de Silva M, et al. The clinical clinical significance of red cell alloantibodies significance of blood group antibodies. Transfus using a monocyte monolayer assay. Transfusion Med 2002;12:287-95. 1987;27: 449-52. 27. Leger RM. In vitro cellular assays and other 38. Garratty G. Predicting the clinical significance of approaches used to predict the clinical red cell antibodies with in vitro cellular assays. significance of red cell alloantibodies: a review. T.ransfus Med Rev 1990;IV:297-312. Immunohematology 2002;18:65-70. 39.Zupanska´ B, Gronkowska A, Ziemski M. 28. Reid ME, Øyen R, Marsh WL. Summary of the Comparison of activity of peripheral monocytes clinical significance of blood group alloantibodies. and splenic macrophages in the monocyte mono- Semin Hematol 2000;37:197-216. layer assay.Vox Sang 1995;68:241-2. 29. Nance S. Review: measuring red cell survival and 40. Nance SJ, Arndt PA, Garratty G.The effect of fresh determining the clinical significance of red cell normal serum on monocyte monolayer assay antibodies. Immunohematology 1995;11:31-8. reactivity.Transfusion 1988;28:398-9. 30. Engelfriet CP, Overbeeke MAM, Dooren MC, 41. Church A, Mallory D, Kavitsky D, Nance S. Impact Ouwehand WH, von dem Borne AEGKr. Bioassays of complement in the monocyte monolayer assay to determine the clinical significance of red cell (abstract).Transfusion 1997;37(Suppl):27S.

alloantibodies based on Fc -induced 42. Branch DR, Gallagher MT. The importance of CO2 destruction of red cells sensitized by IgG. in short-term monocyte-macrophage assays Transfusion 1994;34:617-26. (letter).Transfusion 1985;25:399. 31. Garratty G. Factors affecting the pathogenicity of 43. AuBuchon JP,Brightman A,Anderson HJ,Kim B. An red cell auto- and alloantibodies. In: Nance SJ, ed. example of anti-Yta demonstrating a change in its Immune destruction of red blood cells. Arlington, clinical significance.Vox Sang 1988;55:171-5. VA: American Association of Blood Banks, 1989; 44. DiNapoli J, Gingras A, Diggs E, Alicea-Tossas E, 109-69. Kessler L. Survival of Ge+ red cells in a patient 32. Garratty G. Predicting the clinical significance of with anti-Ge1,2: data from 51Cr, flow cytometric, alloantibodies and determining the in vivo survival IgG subclass, and monocyte erythrophagocytosis of transfused red cells. In: Judd WJ, Barnes A, eds. assays (abstract). Transfusion 1986;26:545. Clinical and serological aspects of transfusion

158 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

Review: all RBCs incompatible—serologic aspects

45. Anderson G, Gray LS, Mintz PD. Red cell survival 60. Sweeney JD, Holme S, McCall L, et al. At(a–) studies with a patient with anti-Tca. Am J Clin phenotype: description of a family and reduced Pathol 1991;95:87-90. survival of At(a+) red cells in a proposita with anti- 46. Nance S, Scandone P, Fassl L, et al. Monocyte Ata. Transfusion 1995;35:63-7. monolayer assay (MMA) results are affected by 61. Baldwin ML, Ness PM, Barrasso C, et al. In vivo transfusion of incompatible red cells (abstract). studies of the long-term 51Cr red cell survival of Transfusion 1997;37(Suppl):37S. serologically incompatible red cell units. 47. Kakaiya R, Sheahan E, Julleis J, et al. 51Chromium Transfusion 1985;25:34-8. studies with an IgG1 anti-Yta (letter). 62. Morel PA, Hamilton HB. Oka: an erythrocytic anti- Immunohematology 1991;7:107. gen of high frequency.Vox Sang 1979;36:182-5. 48.Pearson HA, Richards VL, Wylie BR, et al. 63. Gutgsell NS, Issitt LA, Issitt PD. Transfusion of Assessment of clinical significance of anti-Ge in an antigen-positive, serologically-incompatible blood untransfused man. Transfusion 1991;31:257-9. in immunized patients, based solely on the results 49. Issitt PD, Gutgsell NS, Hervis L. Some stored of a monocyte monolayer assay (MMA) (abstract). antibodies give unreliable results in the monocyte Transfusion 1988;28:32S. monolayer assay (letter). Transfusion 1988;28: 64. Eckrich RJ, Mallory DM, Sandler SG. Correlation of 399-400. monocyte monolayer assays and posttransfusion 50. Wren MR, Issitt PD. The monocyte monolayer survival of Yt(a+) red cells in patients with anti- a assay and in vivo antibody activity (abstract). Yt . Immunohematology 1995;11:81-4. Transfusion 1986;26:548. 65. Zupanska B, Brojer E, McIntosh J, Seyfried H, 51. Gibson M, Devenish A, Daniels GL, Contreras M. Howell P. Correlation of monocyte-monolayer assay results, number of erythrocyte-bound IgG A transfusion problem in a thalassaemic infant molecules, and IgG subclass composition in the with anti-Lu6 (abstract). Ann Mtg Br Blood study of red cell alloantibodies other than D.Vox Transfus Soc 1983;27. Sang 1990;58:276-80. 52. Issitt PD, Valinsky JE, Marsh WL, DiNapoli J, 66. Hildebrandt M, Hell A, Etzel R, Genth R, Salama A. Gutgsell NS. In vivo red cell destruction by anti- Determination and successful transfusion of anti- Lu6.Transfusion 1990;30:258-60. Gerbich-positive red blood cells in a patient with 53. Yahalom V, Ellis MH, Poole J, et al. The rare Lu:-6 a strongly reactive anti-Gerbich antibody. phenotype in Israel and the clinical significance of Infusionsther Transfusionsmed 2000;27:154-6. anti-Lu6.Transfusion 2002;42:247-50. 67. Chapman RL, Hare V. Successful repeated trans- 54. Shirey RS, Øyen R, Heeb KN, Kickler TS, Ness PM. fusion of Cr(a+) blood to a patient with anti-Cra 51 Cr radiolabeled survival studies in a patient with (abstract). Transfusion 1992;32:23S. anti-Lu12 (abstract). Transfusion 1988;28(Suppl): 68. Byrne PC, Eckrich RJ, Malamut DC, Mallory DM, 37S. Sandler SG. Use of the monocyte monolayer assay 55. McSwain B, Robins C. A clinically significant anti- (MMA) to predict the clinical significance of anti- a Cr (letter). Transfusion 1988;28:289-90. Cra (abstract). Transfusion 1995;35:61S. 56. Leatherbarrow MB, Ellisor SS, Collins PA, et al. 69. Kowalski MA, Pierce SR, Edwards RL, et al. a Assessing the clinical significance of anti-Cr and Hemolytic transfusion reaction due to anti-Tca. anti-M in a chronically transfused sickle cell Transfusion 1999;39:948-50. patient. Immunohematology 1988;4:71-4. 70. Kobuszewski M, Wallace M, Moulds M, et al. 57. Bell JA, Johnson ST, Moulds M, et al. Clinical Clinical significance of anti-Lu8 in a patient who significance of anti-Tcab in the second example of received Lu:8 red cells (abstract). Transfusion a Tc(a–b–) individual (abstract). Transfusion 1988;28(Suppl):37S. 1989;29 (Suppl):17S. 71. Bacon J, Sherrin D,Wright RG. Case report, anti-Jra 58. Judd WJ, Oberman HA, Silenieks A, Steiner EA. (letter).Transfusion 1986;26:543-4. Clinical significance of anti-Lan (letter). 72. Kwon MY, Su L, Arndt PA, Garratty G, Blackall DP. Transfusion 1984;24:181. Clinical significance of anti-Jra: report of two cases 59. Ramsey G, Sherman LA, Zimmer AM, et al. Clinical and review of the literature. Transfusion 2004;44: significance of anti-Ata.Vox Sang 1995;69:135-7. 197-201.

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 159 S.T.NANCE AND P.A.ARNDT

73. Leger R, Arndt P, Co A, O’Brien L, Garratty G. Sandra Taddie Nance, MS, MT(ASCP)SBB (correspon- Clinical significance of an anti-Dib assessed by ding author), Director,Technical Services, Penn-Jersey flow cytometry. Immunohematology 1997;13: Region and National Reference Laboratory for Blood 93-6. Group Serology, American Red Cross Blood Services, 74. Nakache R, Levene C, Sela R, Kaufman S, Shapira Z. 700 Spring Garden Street, Philadelphia, PA 19123 Dra (Cromer-related blood group antigen)- and Patricia A. Arndt, MS, MT(ASCP)SBB, Senior incompatible renal transplantation. Vox Sang Research Associate, Southern California Region, 1998;74:106-8. American Red Cross Blood Services, 1130 South 75. Daniels GL, Judd WJ, Moore BPL, et al. A ‘new’ high Vermont Avenue, Los Angeles, CA 90006. frequency antigen Era. Transfusion 1982;22: 189-93. 76. International Committee for Standardization in Haematology. Recommended method for radio- isotope red-cell survival studies. Br J Haematol 1980;45:659-66.

Manuscripts: The editorial staff of Immunohematology welcomes manuscripts pertaining to blood group serology and education for consideration for publication.We are especially interested in case reports, papers on and white cell serology,scientific articles covering original investigations, and papers on the use of computers in the blood bank. Deadlines for receipt of manuscripts for consideration for the March, June, September, and December issues are the first weeks in November, February, May, and August, respectively. Instructions for scientific articles and case reports can be obtained by phoning or faxing a request to Mary H. McGinnniss, Managing Editor, Immunohematology, at (301) 299-7443, or see “Instructions for Authors” in every issue of Immunohematology or on the Web. Include fax and phone numbers and e-mail address with your manuscript.

Free Classified Ads and Announcements Immunohematology will publish classified ads and announcements (SBB schools, meetings, symposia, etc.) without charge. Deadlines for receipt of these items are as follows: Deadlines 1st week in January for the March issue 1st week in April for the June issue 1st week in July for the September issue 1st week in October for the December issue

Fax these items to Mary H. McGinniss, Managing Editor, (301) 299-7443.

160 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 Review: transfusing incompatible RBCs—clinical aspects

G. MENY

Introduction physicians still utilize the Hb level as the “trigger” for “As soon as the blood began to enter into his veins, transfusion. There is no absolute Hb level below which he felt the heat along his arm and under his armpits. a patient will not survive and that makes it difficult His pulse rose and soon after we observed a plentiful when considering transfusing incompatible RBCs. sweat over his face. His pulse varied extremely at this The signs and symptoms experienced by a patient instant and he complained of great in his kidneys, are usually consistent with the rate at which the an- and that he was not well in his stomach, and that he emia developed.2 Weiskopf et al.3 found that acute was ready to choke unless given his liberty. He was isovolemic anemia to a Hb level of < 6 gm/dL resulted made to lie down and fell asleep, and slept all night in mild, reversible changes in memory reaction time, without awakening until morning. When he awakened while < 5 gm/dL impaired immediate and delayed he made a great glass full of urine, of a color as black as memory. Cardiovascular condition and age are also if it had been mixed with the soot of chimneys.”1 important factors to consider when deciding if (and The above description of the signs and symptoms when) transfusion is necessary. Cardiovascular following a blood transfusion were published by Jean compensatory mechanisms to anemia, such as increase Denis, a physician working for King Louis XIV in 1668. in heart rate,increase in stroke volume,and decrease in We can thank Dr.Denis for providing a vivid and classic peripheral vascular resistance, are impaired in patients description of what is now known to be a hemolytic with cardiovascular diseases and (usually) in the 4 transfusion reaction (HTR). Various in vitro (or, less elderly. Carson et al. retrospectively analyzed the 30- frequently, in vivo) immunohematologic techniques day mortality of 1958 surgical patients >18 years of age which attempt to prevent HTRs have evolved over the who refused transfusion for religious reasons. They years. The “goal” is to predict compatibility and trans- compared outcome in those with and without fuse RBCs which will survive and function as well as cardiovascular disease (history of angina, myocardial the recipient’s own RBCs. One is more comfortable infarction, congestive heart failure, or peripheral transfusing “compatible” RBCs. Nonetheless, there are vascular disease). Mortality was 1.3 percent (0.8% to 2.0%) in patients with a preoperative Hb of 12 g/dL or times when “incompatible” RBCs must be transfused. greater and 33.3 percent (18.6% to 51.0%) in patients This paper will review clinical aspects of this decision. with a preoperative Hb < 6 g/dL. A correlation noted between serious morbidity and blood loss in the low Issues for Consideration preoperative Hb group was more apparent in patients Issues that should be considered when caring for a with cardiovascular disease than those without patient requiring incompatible blood are: “Is cardiovascular disease. Hebert et al.5 published a transfusion really necessary?” “Is the antibody(ies) multicenter, randomized, controlled study examining clinically significant?” and “How should the transfusion whether a restrictive or liberal transfusion strategy had be administered?” an effect on mortality in critically ill patients with euvolia. Patients were randomly assigned to maintain When to transfuse incompatible RBCs their Hb to either between 7.0 and 9.0 g/dL (restrictive It can be difficult to state with certainty when to transfusion strategy) or between 10.0 and 12.0 g/dL transfuse incompatible RBCs to an anemic patient as (liberal transfusion strategy). Interestingly, 30-day there are many variables which play a role in mortality rates were similar in the two groups and, in individuals’ responses to their Hb (or Hct) levels. Most fact, were significantly less in the restrictive group in

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 161 G. MENY individuals < 55 years of age and in those with an Acute at http://www.ncbi.nlm.nih.gov/PubMed) can provide Physiology and Chronic Health Evaluation II score of information on general “tendencies” of an antibody’s < 20. This benefit was lost, however, in critically ill clinical significance. Thus, if time is available, in vitro patients with acute myocardial infarction and unstable testing of the patient’s antibody may be desirable and angina. help predict, for example, which RBC units (antigen Tachycardia and postural hypotension, which are positive or antigen negative) should be transfused. The usually present in acute blood loss, are not often seen paper in this issue by Nance and Arndt provides with chronic anemia. A patient with chronic anemia additional information on useful laboratory assays,such may have an increased blood volume due to an as the monocyte monolayer assay (MMA). overexpanded plasma volume. Rapidly administering In vivo testing is rarely performed, although the 1- blood to a patient with severe chronic anemia may hour 51Cr RBC survival study recommended by the precipitate cardiopulmonary decompensation (or International Committee for Standardization in circulatory overload). Indeed, circulatory overload is Hematology is the gold standard for predicting clinical an important and underrecognized complication of significance of a patient’s antibody.9 A radiolabeled transfusion, particularly in the elderly.6 See Popovsky 0.5 mL sample of incompatible RBCs is injected into for a review on this transfusion complication.7 the patient and samples are obtained at 10 and 60 Thus, in summary,while otherwise healthy patients minutes. If radioactivity in the plasma at 10 and 60 usually will require a transfusion when their Hb minutes is < 3% of the total injected and the RBC declines to < ~ 6 to 7 g/dL, these patients may be able survival at 60 minutes is at least 70%, then transfusion to tolerate and compensate at these low Hb levels of the incompatible blood carries minimal risk. Most without transfusion, particularly if the anemia has facilities are not equipped to perform this type of in slowly evolved. However, this does not apply to the vivo testing. patient with cardiovascular disease. Another in vivo test which may be performed is Note that the preceding discussion applies to known as the “in vivo crossmatch” or “biological patients for whom compatible or incompatible RBCs crossmatch.” 10 to 50 mL of unlabeled incompatible are available for transfusion. Decisions to transfuse RBCs is transfused and a sample is collected from the compatible blood in urgent situations are frequently patient posttransfusion to check for hemoglobinemia.10 made with little hesitation. However, transfusing This test only detects intravascular hemolysis. incompatible blood is frequently delayed until the Extravascular RBC destruction cannot be predicted. patient’s life is in serious jeopardy. Transfusion of incompatible blood must occur where there is an The transfusion urgent, life-threatening clinical need, and transfusion If,and when,the decision is made to transfuse,how should not be withheld because of the serologic can the process be made as free of adverse events as findings. possible? It is best to always transfuse using leukocyte- reduced blood components. Although the patient’s Is the antibody clinically significant? clinical condition may not warrant their use, leukocyte- As one is evaluating the patient’s clinical status and reduced blood components can prevent febrile determining the urgency of the RBC transfusion nonhemolytic transfusion reactions, which are requirement, frequently asked simultaneous questions complications of transfusion and can mimic a HTR. For include: “How clinically significant is this RBC example, one does not want to discontinue a RBC antibody?” and “What will happen to the RBCs if transfusion to a patient with multiple alloantibodies transfused—will they be destroyed intravascularly? and autoantibodies because of fever (due to a febrile extravascularly? how rapidly?” Some antibodies are nonhemolytic transfusion reaction). Premedication known to be clinically significant (i.e., cause with antipyretics may mask the fever of a HTR, but hemolysis), such as anti-A, -B, -D, and -K. Others are other signs and symptoms of hemolysis will still be usually not considered clinically significant, such as present. Premedication with an antipyretic or anti-Lea. M.E.Reid and C.Lomas-Francis have written an antihistamine (for patients with a history of allergic excellent source book on blood group antibodies for reactions) may be administered immediately prior to use by transfusionists.8 This book,as well as a literature transfusion if administered intravenously. They should search (e.g.,the National Library of Medicine’s PubMed be given 30 to 60 minutes prior to the start of the

162 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

Review: incompatible transfusion—clinical aspects transfusion if given orally. Some patients may benefit The timing and location of any RBC destruction are from receiving corticosteroids or IVIG (see Case important to note when transfusing incompatible Studies section of this paper). blood. Immune-mediated RBC destruction can be If possible, transfuse the patient during a time categorized by the time of the reaction—acute or when adequately trained staff is available to monitor delayed—and by the location of the hemolysis— the transfusion. This includes laboratory staff available intravascular or extravascular. Acute hemolysis takes to interpret any adverse reactions,should they develop. place within minutes to hours (up to 24 hours after The most knowledgeable individuals for all aspects of a transfusion) and delayed hemolysis occurs within transfusion are usually available during the day, rather several days following transfusion. During intra- than at night. vascular reactions, antigen-antibody interactions result Systems must be in place to select the unit and in binding of complement and generation of the issue and match it to its intended recipient. As with any membrane attack complex (C5b–9). This causes transfusion, make certain that an informed consent osmotic lysis of RBCs, resulting in hemoglobinemia which complies with applicable laws is obtained. Vital and hemoglobinuria. Either IgG or IgM antibodies are signs,including blood pressure,pulse,and temperature, capable of causing intravascular lysis. Intravascular should be recorded prior to initiating the transfusion. hemolysis, for example, could be observed when The patient should be kept well hydrated during the transfusing Vel+ RBCs to a patient with anti-Vel. During transfusion. Once the transfusion is started, the extravascular reactions, the membrane attack complex transfusionist should remain with and observe the is not generated. RBCs become sensitized with IgG patient for at least the first 15 minutes. Vital signs antibodies, complement, or both, and are removed by should be documented at that time. The patient should macrophages within the reticuloendothelial system.12 be observed at frequent intervals. Vital signs should be Extravascular hemolysis, for example, could be recorded at the end of the transfusion. observed when transfusing e+ RBCs to a patient with Davenport summarized the most frequent clinical anti-e. signs and symptoms observed in 90 cases of When a patient has multiple antibodies and intravascular HTRs and 101 cases of extravascular transfusion of incompatible blood is required, HTRs (Table 1).11 Signs and symptoms of an acute HTR incompatible blood is chosen which will survive the frequently become apparent within the first few longest and with minimal adverse effects. For example, minutes of the transfusion, hence the need for the a patient in need of urgent surgery has “antibody 1”and transfusionist to remain with the patient during that “antibody 2.” Blood compatible with both cannot be time. Conscious patients should be educated about obtained without importing it from out of the country possible adverse effects of transfusion and the through the American Rare Donor Program and surgery importance of their immediately reporting any of the must be performed. “Antibody 1” frequently destroys signs or symptoms of a HTR (acute or delayed). If the RBCs intravascularly, while “antibody 2” frequently patient is unconscious, vital signs, such as pulse and destroys RBCs extravascularly. Both antibodies are temperature, should be checked at regular intervals detectable in the patient’s serum. MMAs show that throughout the transfusion and the patient should be both are clinically significant. If compatible blood can observed for evidence of an acute HTR, including be found for only one of the antibodies, which should uncontrollable bleeding (disseminated intravascular be selected? Blood compatible with “antibody 1” coagulation). (incompatible with “antibody 2”) should be selected because otherwise intravascular lysis is likely to occur, Table 1. Signs and symptoms of a HTR at initial presentation11 and it should be avoided. Intravascular HTR Extravascular HTR (% of Patients) (% of Patients) What laboratory tests can be performed after transfusing incompatible blood to assist in the clinical Fever, chills, or both 82 50 management of the patient? Tests which can evaluate 19 13 the presence of both intravascular and extravascular Hypotension, tachycardia, or both 12 — hemolysis (Table 2) should be performed. Many of Nausea, vomiting, or both 12 — these tests are the same as those used in the evaluation Jaundice — 12 of a HTR workup when “compatible” blood is Dyspnea 10 1 transfused. In addition to the Hb and Hct, examination

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 163

G. MENY

Table 2. A diagnostic approach to hemolysis suspected.15 Both hemoglobinemia and hemoglobin- Determine CBC, reticulocyte count, haptoglobin, bilirubin (total uria are rather transient and may be cleared within a and indirect), and urinalysis. Examine peripheral smear. matter of hours. The urine should be examined for • Complete blood count Determine Hb, Hct, and RBC indices hemosiderin if it is suspected that the acute Platelet count usually normal with intra- and extra-vascular intravascular hemolytic episode took place a few days hemolytic anemia previously or is chronic in nature. Hemosiderin is - Thrombocytopenia with positive DAT: Evans syndrome containing granules in the urine which are first shed a • Peripheral smear Spherocytes suggest hemolytic anemia few days after hemolysis begins. Hemosiderin, Help rule out other causes for anemia (e.g., schistocytes, however, may be found in any condition which causes malignancy, hemoglobinopathy, iron deficiency) deposition of iron in the renal parenchyma, such as • Serum haptoglobin Low in acute intravascular hemolysis hemochromatosis or multiple transfusions. Acute-phase reactant Markedly elevated levels of lactate dehydrogenase • Lactate dehydrogenase (LD) are observed with acute intravascular hemolysis Elevated in intravascular hemolysis due to RBC lysis. Although rarely tested for, a LD1:LD2 Elevated in other damaged tissue or neoplastic cells flip may be observed in hemolytic anemia if LD • Serum bilirubin Elevated total and indirect in extravascular hemolysis isoenzymes are examined. The LD1:LD2 flip is also Slight increase in total and indirect in intravascular hemolysis observed in patients with myocardial infarction.16 Higher levels seen in patients with compromised liver function or Gilbert’s disease An elevated reticulocyte count is indicative of but • Urinalysis not specific for hemolysis. Patients with sickle cell dis- Hemoglobinuria seen in intravascular hemolysis ease have a reticulocytosis as a means of compensating Rule out hematuria, myoglobinuria, porphyria, which can also cause “red urine” for shortened RBC survival. If a reticulocytopenia is Hemosiderinuria seen in intravascular hemolysis present and hemolysis is occurring, these patients are at risk for developing life-threatening anemia.17 The of the peripheral blood smear is important.2 For unique challenges of transfusing patients with sickle example, RBCs may appear in aggregates in cold cell disease are briefly discussed later in this paper. agglutinin disease. Spherocytes and microspherocytes Extravascular immune-mediated RBC destruction are usually observed in immune hemolytic anemia. occurs when immunoglobulin- and/or complement- RBC fragments and helmet cells, though, are suggestive sensitized RBCs are destroyed by macrophages within of a microangiopathic hemolytic anemia, not an the reticuloendothelial system. Hemoglobinemia and immune-mediated process. hemoglobinuria typically do not occur. While serum Other laboratory tests can be used to indicate the haptoglobin levels may decrease, they do not reach the presence of intravascular or extravascular hemolysis. very low levels seen with intravascular hemolysis. An acute intravascular hemolytic event tends to be Usually, total serum bilirubin increases only slightly followed by low serum haptoglobin, hemoglobinemia, (from the normal < 1.5 mg/dL to < 6 mg/dL) during a and hemoglobinuria. Langley et al.13 described two hemolytic episode in patients with normal liver patients who developed very low serum haptoglobin function.17 An increase in indirect serum bilirubin and levels which persisted for several days after HTRs. In urine urobilinogen will also be observed. If the ele- contrast, there was no consistent change in vated total serum bilirubin is composed predominantly haptoglobin levels in 21 patients following compatible of indirect bilirubin, hemolysis or Gilbert’s disease is blood transfusion. Haptoglobin is an acute-phase likely to be present.14 reactant and comorbid conditions such as infection may mask the diagnosis by somewhat elevating Case Studies haptoglobin levels.14 However, in classic acute In a patient with multiple antibodies or an antibody intravascular hemolysis, the haptoglobin level is usually to a high-frequency antigen, one may anticipate the < 25 mg/dL and may approach 0 mg/dL. need to transfuse “incompatible” blood. The following Hemoglobinemia can be detected by inspecting a two case studies, and a paper in this issue by A.M. sample of the patient’s plasma. A centrifuged urine Svensson et al., illustrate how a decision is reached. sample is examined for hemoglobinuria. Care must be A 77-year-old woman with a 6.7 g/dL Hb required taken to differentiate intact RBCs from free transfusion (i.e., she was clinically symptomatic), but hemoglobin and free myoglobin, if rhabdomyolysis is no compatible blood was available in the United States.

164 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

Review: incompatible transfusion—clinical aspects

She was group O, D–, with anti-hrB, -E, and -S. MMAs successfully treated,at age 15,and pregnancy,at age 19. were performed by the American Red Cross National RBC alloantibodies previously identified included anti- Reference Laboratory for Blood Group Serology and Fya, -S, and -Leb. She received two units of group O, D–, the results were interpreted as “clinically significant,” Fya–, S–, Leb– RBCs and was discharged. Several days (i.e., these antibodies would result in less than normal later, she was readmitted with declining Hb (4.5 g/dL). RBC survival). Blood was available from another She was transfused with one unit of group O, D–, Fya–, country, but it would take several days to arrive. S–, Leb– RBCs, but her Hb declined the next day to 3.1 How could this patient be managed until g/dL. Additional antibodies could not be identified as compatible blood was available? Initially, it was noted an explanation for the apparent HTR. While one may that RBCs negative for her “formed”antibodies, but D+, feel the need to continue transfusing this patient, this were available. Two units of D+, hrB–, E–, S– RBCs were case illustrates many of the features of the “sickle cell transfused and her Hb increased to 10 g/dL. However, hemolytic transfusion reaction,”i.e., manifestation of a as an anti-D developed, the Hb declined to 5.1 g/dL delayed HTR, development of a more severe anemia over the next 6 days. Three units of “incompatible”D–, after transfusion than was present before, and E–, S–, hrB+ RBCs were then transfused, raising the subsequent transfusions that further exacerbate patient’s Hb to 8.6 g/dL. These units were destroyed anemia that may become life-threatening; both RBC extravascularly,and Hb declined to 6.7 g/dL over a few alloantibodies and autoantibodies can be present; days until compatible blood arrived from South Africa. serologic studies may not explain the HTR; and Two units of group O, D–, hrB–, E–, S– RBCs were administration of corticosteroids may lead to a gradual 18–20 transfused and the Hb increased to 9.6 g/dL, where it improvement in some patients’ Hb. This patient’s remained stable until the patient was discharged. Hb improved to 7.8 g/dL after receiving 60 mg/day of To summarize this patient’s transfusion manage- prednisone without additional transfusion. In a case 20 ment, multiple alloantibodies were identified. Initially, report described by Petz et al., their patient with a b time was available, which permitted performing an in sickle cell anemia and anti-E, -C, -K, -S, -Fy , and -Jk vitro assay (MMA) to assist in predicting the clinical received a total of 15 units of RBCs and her Hct significance of the patient’s antibodies. Her clinical dropped from 13.2% to 9.3%. Prednisone (60 mg/day) was started, two units of RBCs were transfused,and the condition dictated that transfusion be performed. This Hct increased to 22.4% prior to discharge 1-week later. is important—transfusion should never be withheld While this patient responded to steroid medication, from a patient with a clinical need based on a serologic consideration can be given to also treating with IVIG incompatibility. Until compatible units could be to prevent RBC destruction.21 located, D+ but otherwise-compatible RBCs were transfused (which were eventually destroyed by the formation of anti-D), followed by transfusion of Summary incompatible D–, E–, S–, hrB+ RBCs (also eventually As with any transfusion, the risks must be weighed destroyed via extravascular mechanisms). Both sets of against the benefits for each individual patient prior to transfusions permitted proper clinical management of transfusion. Transfusion should not be withheld from the patient without serious morbidity and mortality the patient with an urgent, life-threatening clinical that could have occurred while awaiting arrival of need on the basis of serologic test results. If incom- compatible blood. patible blood must be transfused, it is important for While the above case illustrates how one may clinical care staff and laboratory staff to communicate transfuse against different “incompatibilities” until frequently to determine which RBC units should be compatible blood arrives, this approach may not work selected for transfusion, when the transfusion will be well in every clinical situation. One such scenario to administered, and how the patient will be monitored. keep in mind is the patient with sickle cell disease who is developing a progressively more severe anemia with References each transfusion. The following case illustrates this 1. Oberman HA. The history of transfusion point. medicine. In: Petz LD, Swisher SN, Kleinman S, A 21-year-old woman with sickle cell anemia was Spence RK, Strauss RG. Clinical practice of admitted with in December. Her medical transfusion medicine. 3rd ed. New York: Churchill history is significant for Burkitt’s lymphoma, Livingstone, 1996:11-32.

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 165 G. MENY

2. Hoffman R, Benz EJ Jr, Shattil SJ, et al., eds. 13. Langley GR, Owen JA, Padanyi R. The effect of Hematology basic principles and practice. 3rd ed. blood transfusion on serum haptoglobin. Br J New York: Churchill Livingstone, 2000:367-75. Haematol 1962;8:392-9. 3. Weiskopf RB, Kramer JH, Viele M, et al. Acute 14. The diagnosis of hemolytic anemias. In: Petz LD, severe isovolemic anemia impairs cognitive Garratty G. Immune hemolytic anemias. 2nd ed. function and memory in humans. Anesthesiology Philadelphia: Churchill Livingstone, 2004:33-60. 2000;92:1646-52. 15. Trainor LD, Solomon HM. Detecting myoglobin- 4. Carson JL, Duff A, Poses RM, et al. Effect of uria: a low-tech analysis. Lab Med 1997;28:569-71. anaemia and cardiovascular disease on surgical 16. Jacobs DS, Garg U, Oxley DK. Chemistry.In: Jacobs mortality and morbidity. Lancet 1996;348: DS, Oxley DK, DeMott WR, eds. Laboratory test 1055-60. handbook.5th ed.Hudson,OH:Lexi-Comp,75-302. 5. Hebert PC, Wells G, Blajchman MA, et al. A 17. Petz LD, Calhoun L, Shulman IA, et al. The sickle multicenter,randomized,controlled clinical trial of cell hemolytic transfusion reaction syndrome. transfusion requirements in critical care. N Engl J Med 1999;340:409-17. Transfusion 1997;37:382-92. 6.Popovsky MA, Audet AM, Andrzejewski C. 18.Cullis JO, Win N, Dudley JM, Kaye T. Post- Transfusion-associated circulatory overload in transfusion hyperhaemolysis in a patient with orthopedic surgery patients: A multi-institutional sickle cell disease: use of steroids and intravenous study. Immunohematology 1996;12:87-9. immunoglobulin to prevent further red cell 7. Popovsky MA. Circulatory overload. In: Popovsky destruction.Vox Sang 1995;69:355-7. MA, ed. Transfusion reactions. 2nd ed. Bethesda, 19.King KE, Shirley RS, Lankiewicz MW, Young- MD: American Association of Blood Banks, 2001: Ramsaran J, Ness PM. Delayed hemolytic 255-60. transfusion reactions in sickle cell disease: 8.Reid ME, Lomas-Francis C. The blood group simultaneous destruction of recipients’ red cells. antigen factsbook. 2nd ed. San Diego: Academic Transfusion 1997;37:376-81. Press, 2003. 20. Petz LD,Calhoun L,Shulman IA,Johnson C,Herron 9. International Committee for Standardization in RM. The sickle cell hemolytic transfusion reaction Hematology. Recommended method for radio- syndrome. Transfusion 1997;37:382-92. isotope red-cell survival studies. Br J Haematol 21. Cullis JO, Win N, Dudley JM, Kaye T. Post- 1980;45:659-66. transfusion hyperhaemolysis in a patient with 10. Mollison PL. Survival in vivo as a test for red cell sickle cell disease: use of steroids and intravenous compatibility. Haematologia 1972;6:139-45. immunoglobulin to prevent further red cell 11. Davenport RD.Hemolytic transfusion reactions.In: destruction.Vox Sang 1995;69:355-7. Popovsky MA, ed. Transfusion reactions. 2nd ed. Bethesda, MD: American Association of Blood Geralyn Meny, MD, American Red Cross Blood Banks, 2001:1-44. Services, 700 Spring Garden Street, Philadelphia, PA 12. Mechanisms of immune hemolysis. In: Petz LD, 19123 Garratty G. Immune hemolytic anemias. 2nd ed. Philadelphia: Churchill Livingstone, 2004:133-65.

REMEMBER: THE PASSWORD IS “2000” For www.redcross.org/pubs/immuno Now, as a subscriber, you can enter the password, 2000, and access the back issues. That means cover to cover! You will receive every article in total, every letter to the editor, every review, every ad, every notice, and every literature review! All of the other services will continue to be available on the Web site, including sending a letter to the editor, subscribing with a credit card on the secure order site, performing a literature search, reviewing instructions for authors, and linking to other important sites. Log on now to see this great service!

166 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 Review: evaluation of patients with immune hemolysis

LAWRENCE D. PETZ

The presence of hemolysis is usually easily Table 1. Differential diagnosis of immune hemolytic anemias confirmed by simple tests such as reticulocyte count, Autoimmune hemolytic anemias (AIHA) review of the peripheral blood film, serum bilirubin Warm-antibody AIHA 1 Idiopathic (direct and indirect), LDH, and haptoglobin. When it Secondary (e.g., chronic lymphocytic leukemia, lymphomas, has been established that the patient has a hemolytic systemic lupus erythematosus) anemia, the cause of the hemolysis should be sought Cold agglutinin syndrome next. The DAT should be performed on the RBCs of Idiopathic Secondary every patient in whom the presence of hemolysis has Nonmalignant disorders (e.g., mycoplasma pneumoniae been established, and a positive DAT in a patient with infection, infectious mononucleosis, other virus infections) Malignant disorders (e.g., lymphoproliferative disorders) hemolytic anemia does, of course, indicate that the Combined cold and warm AIHA most likely diagnosis is one of the immune hemolytic 2 Paroxysmal cold hemoglobinuria anemias (IHAs). However, there are numerous causes Idiopathic of IHA and the clinical manifestations, prognosis, and Secondary Viral syndromes therapy vary so that defining the precise diagnosis is of Syphilis considerable importance. Atypical AIHA AIHA with a negative direct antiglobulin test Classification of IHAs Warm-antibody AIHA caused by IgM or IgA autoantibodies Drug-induced immune hemolytic anemia In evaluating a patient with IHA, one should first Drug-related antibody identifiable consider a differential diagnosis as outlined in Table 1. Drug-induced AIHA Although a definitive diagnosis rests with serologic Alloantibody-induced immune hemolytic anemia Hemolytic transfusion reactions studies, some clinical and routine laboratory pro- Hemolytic disease of the fetus and newborn cedures are sufficiently distinctive as to strongly Note: Tables 1, 2, 4, 5, and 6 were taken from Petz LD, Garraty G. Immune hemolytic suggest the type of IHA that is present. anemias. 2nd ed. Philadelphia: Churchhill Livingstone, 2004.1

Distinctive Clinical Signs and Routine (e.g., in childhood); is rarely paroxysmal; is only Laboratory Procedures occasionally clearly precipitated by cold; and is not invariably expressed as hemoglobinuria, although the Association with exposure to cold latter finding is very common.4 A history of acrocyanosis and/or hemoglobinuria on exposure to cold in an elderly patient with an Autoagglutination acquired hemolytic anemia strongly suggests a Autoagglutination is a finding that may be noted by diagnosis of cold agglutinin syndrome (CAS). However, technologists in all sections of the laboratory, not just these manifestations are absent in a majority of patients those in the blood transfusion or immunohematology with CAS even though they were often emphasized in laboratories. Indeed, cold autoagglutinins that react the early medical literature. strongly at room temperature cause such striking Although one might assume that paroxysmal cold findings that they are difficult to ignore. Autoagglu- hemoglobinuria (PCH) is commonly precipitated by tination visible to the naked eye occurring at room exposure to cold, this is only occasionally true. Indeed temperature is characteristic of the CAS, but may also Wolach et al.3 pointed out that the most common form be noted in about one-third of patients with warm of PCH is the transient type, secondary to infection autoimmune hemolytic anemia (WAIHA).1 Although

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 167 L.D. PETZ the autoagglutination caused by cold agglutinins is Hemoglobinemia and hemoglobinuria often 2+ to 4+, it almost always completely disperses Hemoglobinuria (Hb in the urine) is far less after a few minutes of incubation at 37°C, whereas that common than hematuria (RBCs in the urine), and a caused by warm autoantibodies is usually much common clinical error is the assumption that a weaker and will not disperse at 37°C. If the blood patient’s red urine is caused by hematuria. It should be sample has been obtained from a patient known to remembered that hemoglobinuria, associated with have hemolytic anemia, such simple observations offer hemolytic anemia, cannot occur without hemoglobin- an important clue to the correct diagnosis. emia. If red urine is present without hemoglobinemia, However,a common error is the over-interpretation it should be suspected that the cause is hematuria and of cold agglutination. Many cold antibodies are not hemolysis. The presence of hemoglobinemia and reactive at room temperature but are clinically benign, hemoglobinuria should alert the clinician to a specific albeit somewhat a nuisance in the laboratory. The group of diagnoses and, when considered in association with the clinical setting, often makes the serologic criteria for distinguishing clinically benign specific diagnosis evident (Table 2). Probably the most cold agglutinins from pathologic cold agglutinins common associated diagnoses are hemolytic trans- capable of causing a CAS are discussed later. fusion reactions and severe acute WAIHA, although hemoglobinuria may occur in patients with CAS, Drug ingestion especially after exposure to cold. Also, drug-induced A temporal history of drug ingestion may suggest IHAs caused by cefotetan and ceftriaxone are the etiology of the patient’s IHA. A critical aspect of commonly associated with hemoglobinemia and evaluation of a patient with IHA is the elicitation of a hemoglobinuria. history of drug ingestion which,in some instances,may Table 2. have occurred a week or more prior to the onset of Causes of hemoglobinuria Acute Hemoglobinuria hemolysis or be a single dose given for surgery (e.g., Incompatible blood transfusion cefotetan). Knowledge of the drugs that have been Transfusion of damaged blood (overheating or freezing, bacterial contamination, pump-oxygenation) implicated as a cause of drug-induced IHA is essential. Drugs and chemical agents (immune or nonimmune mechanisms) Many drug-induced IHAs can be distinguished from Paroxysmal cold hemoglobinuria Acute severe warm-antibody AIHA autoimmune hemolytic anemia (AIHA) by laboratory perfringens infection findings, i.e., the demonstration of a drug-dependent Malaria (blackwater fever) Bartonellosis, babesiosis, , toxoplasmosis RBC antibody. However, the administration of some Peritoneal hemorrhage drugs causes hemolytic anemia that is serologically Severe hypophosphatemia Snake and spider bites indistinguishable from cases of idiopathic WAIHA. Cold agglutinin syndrome* Such cases are appropriately termed drug-induced March hemoglobinuria Microangiopathic hemolytic anemia AIHA, whereas those cases wherein a drug-dependent Hypotonic bladder irrigation during prostatic surgery antibody can be identified are termed drug-induced Mistaken intravenous administration of IHA. Chronic Hemoglobinuria Paroxysmal nocturnal hemoglobinuria** Prosthetic cardiovascular materials Alloantibody-induced IHA *Chronic low grade intravascular hemolysis is common, with acute hemoglobinuria Alloantibody-induced hemolytic anemias include resulting from exposure to cold **Characteristically associated with intermittent episodes of grossly evident HDN and hemolytic transfusion reactions. The clinical hemoglobinuria setting usually strongly suggests these diagnoses. Hemoglobinuria and hemoglobinemia are much Although autoantibody-induced HDN can occur as a more common in children, and both warm and cold result of transplacental passage of a mother’s IgG warm AIHA can be the cause. In a child, PCH should be autoantibody, this is very unlikely unless the mother strongly suspected and a Donath-Landsteiner (DL) test has obvious and quite severe WAIHA. should be performed whenever an acute severe Also, when hemolysis occurs in the immediate hemolytic anemia occurs with hemoglobinemia and aftermath of a RBC transfusion, the diagnosis of an hemoglobinuria. Indeed, hemoglobinuria is a common acute hemolytic transfusion reaction is quite evident. presenting manifestation of PCH4 and therefore However, distinguishing a delayed hemolytic trans- becomes an important diagnostic clue. fusion reaction from AIHA is, on occasion, difficult.5–7

168 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

Review: evaluation of immune hemolysis

RBC morphology and erythrophagocytosis Table 3. Secondary autoimmune hemolytic anemias One easily performed and very valuable test for Ovarian tumors Ulcerative colitis determining the specific diagnosis in a patient with Chronic lymphocytic leukemia hemolytic anemia is examination of the peripheral Hodgkins disease Non-Hodgkins lymphoma blood film. RBC morphology often strongly suggests a Other lymphoproliferative disorders specific diagnosis or a limited number of diagnostic Systemic lupus erythematosus (SLE) possibilities. Spherocytosis may be a prominent feature Antiphospholipid syndrome Collagen disorders other than SLE in IHAs, especially WAIHA, ABO HDN, hemolytic Thymoma transfusion reactions, and some instances of drug- Carcinomas Primary immunodeficiency diseases induced hemolysis. Autoimmune lymphoproliferative syndrome (ALPS) Less well appreciated is the fact that RBC Miscellaneous disorders adherence and erythrophagocytosis by neutrophils is a AIHA associated with infectious agents Mycoplasma pneumoniae prominent finding in PCH, but is seen rarely in other Infectious mononucleosis (Epstein-Barr Virus) forms of IHA. Erythrophagocytosis is rarely observed Cytomegalovirus (CMV) Human immunodeficiency virus (HIV) in the peripheral blood film of WAIHA and when it is Varicella (chickenpox) observed, monocytes, not neutrophils, are more often Rubella Parvovirus B19 involved. Hepatitis Malaria and other blood parasites Association with Mycoplasma pneumoniae Bacterial infections If a patient with Mycoplasma pneumoniae infection develops AIHA, the diagnosis of a CAS must relationship between immunologic disorders, includ- be strongly suspected, since AIHA in this setting is ing immune deficiency states, and AIHA is strong.15–19 almost always caused by cold agglutinins. Although the pathogenetic basis for the association between cytopenias and congenital immune deficiency Association with underlying disorders is unclear, defects in T-cell regulation, cytokine defects, (secondary IHAs) abnormal apoptosis, and abnormal production of Patients should be evaluated for certain disease immunoglobulins with autoimmune features are entities, since approximately half of AIHAs are potential mechanisms.15 associated with an underlying disorder (Table 3). AIHAs are classified as secondary for any of several Laboratory Diagnosis of IHAs reasons. One reason is the association of AIHA with an Even in the presence of valuable clinical clues that underlying disease with a frequency greater than can may suggest a specific diagnosis, the confirmation of be explained by chance alone, as in chronic lympho- the precise diagnosis of the type of IHA present cytic leukemia or systemic lupus erythematosus. depends on the laboratory. The serologic tests to be Another criterion for categorizing a given case of performed determine whether the patient’s RBCs are AIHA as secondary is the reversal of the hemolytic coated with IgG, complement components, or both. anemia simultaneously with the correction of the The performance of the DAT supplies such infor- associated disease. Ovarian tumors are a good ex- mation. Further tests must be performed to determine ample;well-documented cases of cure of the AIHA after the characteristics of the antibodies in the patient’s surgical removal of the tumor have been reported.8–11 serum and in a RBC eluate. Similarly, AIHA in association with ulcerative colitis almost invariably goes into remission after colectomy, Significance of the DAT in the diagnosis of IHAs even when hemolysis is refractory to other therapeutic The DAT, using polyspecific and monospecific approaches.12–14 antiglobulin reagents, provides useful information in Still another reason for suspecting a relationship the evaluation of a patient with IHA. However, the between the occurrence of AIHA and an associated results must always be interpreted in conjunction with disease consists of evidence of immunologic aberration clinical and other laboratory data to avoid erroneous as part of the underlying disorder, especially if the conclusions. It must be remembered that a positive associated disease is thought to have an autoimmune DAT occurs in situations other than IHAs. A positive pathogenesis. In general, the evidence for a DAT does not necessarily indicate the presence of

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 169

L.D. PETZ autoantibody; furthermore, even if autoantibody is group O RBCs and autologous RBCs at 20°C (room present, the patient may or may not have a hemolytic temperature can be used) and 37°C (all stages strictly anemia. Thus, an independent assessment must be at 37°C). A duplicate set of tubes is used with added made to determine the presence or absence of fresh complement (pooled normal sera) at optimal pH hemolytic anemia, and the role of the DAT is to aid in (6.5 to 6.8) for lysis. After incubation, the tubes are the evaluation of the etiology of hemolysis when inspected for hemolysis, agglutination, and sensitiz- present. The results of DATs in patients with various ation (antiglobulin test using polyspecific antiglobulin types of IHAs are indicated in Table 4. serum). The results of this screen usually indicate whether Table 4. Typical DAT results in patients with immune hemolytic anemias one is dealing with a cold autoagglutinin, a “warm” Causes of hemolytic anemia Anti-IgG Anti-C3* autoantibody,or possibly a combination of both. Other Warm-antibody AIHA points of interest are whether there is a hemolysin 67% + + 20% +0present and whether it is a “cold”or “warm”hemolysin. 13% 0+If agglutination occurs at 20°C, a cold agglutinin titer Warm-antibody AIHA associated with and thermal amplitude should be performed. Results systemic lupus erythematosus + + of the AIHA serum screen in WAIHA and in CAS are Cold agglutinin syndrome (100%)* 0 + indicated in Tables 5 and 6, respectively. Paroxysmal cold hemoglobinuria (100%) 0+ With the results of the DAT, AIHA screen, and Drug-induced immune hemolytic anemias possibly a cold agglutinin titer/thermal amplitude, one Drug-dependent antibodies Penicillin and first-generation cephalosporins + (+)† generally has a good idea of whether the diagnosis is Second- and third-generation cephalosporins + + WAIHA or CAS. Sometimes the patient’s history and Associated with “Immune Complex Mechanism” + + results of the DAT/AIHA screen will lead to further Drug-independent antibodies + (+) tests, such as the DL test for PCH, or detection of drug- Drug-induced nonimmunologic adsorption of proteins* + (+) dependent antibodies. Hemolytic disease of fetus/newborn + 0 Hemolytic transfusion reactions + (+) Characteristic serology of WAIHA *IgG +C3 were both detected on the RBCs of one patient with CAS because the Autoantibodies causing WAIHA are usually IgG but patient was on methyldopa therapy and made IgG autoantibodies can be IgM or IgA. These proteins can be present †(+) = sometimes positive together; it is rare for only IgM or IgA to be the cause of AIHA. Autoantibodies are usually only found in the AIHA associated with warm IgM autoantibodies serum when all autoantigens are saturated, apparently without the presence of IgG occurs on rare occasions. because the patient’s RBCs are adsorbing warm However, even with potent antisera, RBC-bound IgM is autoantibodies continuously. Only about 60 percent of 1,20,21 difficult to detect with the antiglobulin test. patients’ sera will react with saline-suspended RBCs, Fortunately, IgM antibodies that cause IHA character- istically fix complement, which is much more readily Table 5. Results of serum screening in 244 patients with warm-antibody detected. IgA antibodies only infrequently play a role AIHA Acidified serum in RBC sensitization, and in such cases other immune Untreated plus acidified globulins and/or complement components are almost Results serum complement always, although not invariably, found on the cell Serologic reactions (% Positive reactions) (% Positive reactions) surface as well. The clinical and hematologic features 20°C Untreated RBCs of WAIHA associated only with IgA autoantibodies are Lysis 0.4 0.8 Agglutination 34.8 34.8 very similar to AIHA associated with warm IgG 20°C Enzyme-treated RBCs autoantibodies. Lysis 1.6 2.5 Agglutination 78.6 78.6 The AIHA serum screen test 37°C Untreated RBCs Lysis 0.4 0.4 The “AIHA screen” is an elaboration of the routine Agglutination 4.9 4.9 antibody screen performed for pretransfusion testing. Indirect anti-globulin test 57.4 57.4 In summary,the patient’s serum is tested against a pool 37°C Enzyme-treated RBCs of untreated and enzyme (e.g., ficin)-treated allogeneic Lysis 8.6 12.7 Agglutination 88.9 88.9

170 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

Review: evaluation of immune hemolysis

Table 6. Results of serum screening in 57 patients with CAS insignificant, albeit abnormal, cold agglutinins; has Acidified serum WAIHA with an associated but probably insignificant Untreated plus acidified Results serum complement elevation of cold agglutinin titer and/or thermal Serologic reactions (% Positive reactions) (% Positive reactions) amplitude; or has CAS. A rather common diagnostic 20°C Untreated RBCs error is over-diagnosis of CAS in patients who have Lysis 2.0 14.3 benign cold antibodies and pathogenic warm Agglutination 98 98 autoantibodies. Also, in rare patients, characteristic 20°C Enzyme-treated RBCs findings of both WAIHA and CAS occur simultaneously. Lysis 24.5 93.8 Agglutination 100 100 A diagnosis of CAS must be considered in all 37°C Untreated RBCs patients with acquired hemolytic anemia who have a Lysis 0 0 positive DAT using anti-C3 and a negative DAT using Agglutination 10.7 10.7 Indirect anti-globulin test 5.4 5.4 anti-IgG. A practical initial serum screening procedure 37°C Enzyme-treated RBCs is to test the ability of the patient’s serum to agglutinate Lysis 12.2 22.5 saline-suspended normal RBCs at 20°C (or room Agglutination 28.6 28.6 temperature) after incubation for 30 to 60 minutes. If this screening test is negative, a CAS is extremely but a higher percentage will react in the presence of unlikely; if positive, further studies are necessary to potentiators (e.g., PEG) or enzyme-treated RBCs. determine the thermal amplitude of the antibody. Some sera contain warm hemolysins, which react When CAS appears to be a possible diagnosis on optimally at 37°C but may react at 20°C. The hemolysis the basis of the preceding evaluation, studies of the is enhanced by adding fresh complement at pH 6.5 to thermal range of reactivity of the antibody in saline and 22 6.8 to the patients’ sera. von dem Borne et al. showed albumin are indicated. It is also convenient to simul- that such hemolysins were usually IgM autoantibodies; taneously determine possible Ii blood group specificity they were maximally reactive at a pH of 6.5 and 7 of 11 of the antibody. The titer of the cold agglutinin in CAS (64%) reacted optimally at 30°C; 4 of 11 reacted is invariably highest at 4°C and progressively decreases optimally at 37°C. About one-third of sera from at higher temperatures. Of particular note are the patients with WAIHA contain IgM cold autoagglutinins reactions at 30°C and 37°C. If the reactions at 30°C are that can react quite strongly at 20°C (or room positive in saline or albumin, the antibody may well be temperature), but have a normal cold agglutinin titer at of pathogenic significance, i.e., it may be causing short 4°C and do not react at 30°C. These may be naturally RBC survival in vivo. If the reactions at 37°C are also occurring cold antibodies that become boosted (e.g., positive in the presence of albumin (as is true in about raised thermal amplitude) during the pathogenic 68% of patients) or even when albumin is not present autoimmune response. (only about 7% of patients), the antibody will cause A diagnosis can usually be reached on the basis of problems in compatibility testing. the serologic tests described thus far. In spite of the Using clinical information, the results of the DAT, seemingly complicated nature of the foregoing, the and the preceding screening tests, a reasonably usual or “typical”essential diagnostic tests that lead to confident assessment of the presence or absence of a reasonably confident diagnosis of WAIHA may be very CAS may be made. CAS may be diagnosed if the simply summarized as follows: (a) the presence of an following are present: (1) clinical evidence of acquired acquired hemolytic anemia, (b) a positive DAT, and (c) hemolytic anemia, (2) a positive DAT caused by an unexpected antibody in the serum and eluate that sensitization with C3,(3) a negative DAT using anti-IgG, reacts optimally at 37°C. The antibody usually reacts (4) the presence of a cold autoagglutinin with with all normal erythrocytes but, in some cases, it can reactivity up to at least 30°C in saline or albumin, and readily be shown to react preferentially with antigens (5) a cold agglutinin titer (at 4°C) ≥ 256, except in on the patient’s own RBCs. exceptional cases. An alternative diagnosis must be sought for patients who do not satisfy all these criteria. Characterization of antibodies in CAS The mere presence of cold autoagglutinins is not Patients who have warm and cold autoantibodies diagnostic of CAS, and when they are encountered, the Approximately one-third of WAIHA patients have task is to determine whether the patient has clinically cold agglutinins that can react quite strongly at room

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 171

L.D. PETZ temperature but have a normal titer (at 4°C) and do not RBCs are washed with ice-cold saline and ice-cold react at 30°C and 37°C. Such antibodies are probably antiglobulin serum is used. Indeed, Dacie found that not pathogenic. However, some patients with WAIHA the IAT was a more sensitive way of demonstrating have cold antibodies that react up to 30°C or above and antibody activity than looking for lysis.32 Such therefore may be pathogenic. Some of these patients agglutination tests must be carefully controlled have IgG and C3 on their RBCs, and their sera contain because many sera give positive results under these IgG 37°C-reactive antibodies together with high-titer, circumstances if a broad-spectrum antiglobulin serum high-thermal amplitude cold autoagglutinins (i.e., the is used, due to the presence in human sera of normal combined serology of classical WAIHA and CAS).1,23–28 incomplete cold antibody. It is important, therefore, to Other patients have IgG and/or C3 on their RBCs, and be sure that monospecific anti-IgG antiglobulin serum their sera contain IgG 37°C-reactive antibodies is used. together with cold autoagglutinins of normal titer that Since PCH is quite rare,one may justifiably question react at 37°C and/or 30°C. There are only three the advisability of performing a DL test routinely in published reports that relate to this group, and the patients with acquired hemolytic anemia. However, report by Sokol et al.28 does not give any cold one should be liberal with the indications for agglutinin titers, so some of the patients may belong to performance of the test, since it is simple to perform the first group. Shulman et al.27 believe that up to 8 and its inclusion avoids diagnostic errors. The perfor- percent of WAIHAs may belong to this group. mance of the test is indicated in any child, any patient with hemoglobinuria, patients with a history of PCH hemolysis exacerbated by cold, and in all cases with The diagnosis of PCH or the exclusion of that “atypical” serologic findings. If positive results are diagnosis in the laboratory is usually considerably obtained in the DL test,determination of the specificity easier than that of either WAIHA or CAS. The essential of the autoantibody is indicated. In almost all cases of laboratory test is the DL test. A negative test excludes PCH the antibody has anti-P specificity. Sokol et al.33 the diagnosis of PCH and a positive test is, with rare found anti-P specificity in 27 of 30 (90%) patients with exceptions (described below), diagnostic of the PCH; specificity was not clearly defined in the other disorder. three cases. Nevertheless, rare reports of other The autoantibody associated with PCH is termed a specificities said to be associated with PCH have been biphasic hemolysin, that is to say, it sensitizes RBCs in reported. RBCs necessary for determining anti-P the cold but only hemolyzes them when the RBCs specificity are rare but, with the assistance of reference reach 37°C. The diagnostic test is the DL test, wherein laboratories, specificity testing can be carried out. It RBCs are incubated with the patient’s serum at 0°C should be noted that the DL test must be used to (e.g.,melting ice) and then moved to 37°C for a further determine specificity. incubation. No lysis occurs following the incubation at 0°C, and no lysis occurs if the incubation is carried out Cautions regarding the interpretation of the DL test only at 37°C. The thermal amplitude of this antibody is The DL test is essentially diagnostic for PCH, but usually less than 20°C, that is to say, the antibody will one must be cautious when using serum from patients give a positive DL test only when the initial incubation with CAS. This is true because about 15 percent of sera is < 20°C;stronger results will occur as the temperature from patients with CAS contain monophasic cold of the initial incubation is lowered. Rare patients have hemolysins that will hemolyze untreated RBCs at been described, in whom the DL test is positive when around 20°C. Up to 95 percent of such sera will cause the first incubation phase is as high as 32°C,or their DL direct lysis of enzyme-treated RBCs at 20°C. During the antibody sensitized RBCs up to 37°C, as detected by performance of the DL test, there is a brief period of the IAT.29–31 time when cells and serum are at room temperature The autoantibody may sometimes agglutinate RBCs after being moved from the ice bath to a 37°C water in addition to giving a positive DL test.1 The bath. In about 15 percent of patients with CAS, falsely agglutination is usually of low titer (< 64) at 4°C and of positive (weak) DL tests will occur on this basis. If low thermal amplitude (< 20°C). The DL antibody is enzyme-treated RBCs are used for the DL test, as IgG but is usually only detectable by IAT if, following suggested by some investigators,4 the falsely positive incubation of the patient’s serum and RBCs at 0°C, the rate is likely to be higher. Therefore, if enzyme-treated

172 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

Review: evaluation of immune hemolysis

RBCs are used for the DL test,a control for monophasic between a DHTR and AIHA. The following measures lysis set up in parallel should be mandatory. will afford important clues.

Performing the DL test in patients with Comparison of DAT and IAT hemoglobinemia A simple observation that may yield valuable It may be impossible to determine if in vitro information is the comparison of the strength of the hemolysis has occurred in the DL test if the patient’s DAT and the IAT. In WAIHA, the DAT is almost always serum is red because of marked hemoglobinemia. In stronger than the IAT. It appears that autoantibody is this case, a simple procedure is to perform the cold largely adsorbed onto the patient’s RBCs, and only phase of the DL test using the patient’s serum,but after when the RBCs are heavily coated does one find a large incubation at 4°C, carefully replace the patient’s serum amount of antibody in the serum. In contrast, strongly with fresh normal serum before moving to the 37°C reactive alloantibodies may be present in a patient’s phase of the test. As a control, a similar tube can be serum, but this finding cannot result in a strongly kept at 4°C, or a similar test can be performed with P- positive DAT unless large numbers of transfused RBCs negative RBCs, if available, and if the specificity of the of appropriate antigenic type are present. Thus, the DL antibody is anti-P. Another simple technical aid is to presence of a weakly positive DAT in association with compare the size of the RBC buttons following a strongly positive IAT is presumptive evidence for the centrifugation. If these approaches fail, then a “cold presence of an alloantibody. These findings are IAT” can be performed. therefore highly suggestive of a DHTR. Further, even if the DAT is strongly positive at the time of initial Differentiating Delayed Hemolytic evaluation, it may soon become weaker in subsequent Transfusion Reactions From Autoimmune tests as a result of the destruction of the transfused Hemolytic Anemia RBCs. This is true even though the DAT may remain weakly positive for several months following a DHTR. Delayed hemolytic transfusion reactions (DHTRs) In contrast, a rapid diminution in the strength of the are a recognized risk of blood transfusion. The reaction DAT would not be expected in AIHA except as a result is caused by the reappearance of an antibody, of treatment, as with corticosteroids, immuno- presumably first stimulated by pregnancy or a previous suppressive drugs, or splenectomy. transfusion. Unlike immediate transfusion reactions, which are usually caused by human error,34–38 delayed Antibody specificity reactions are usually not avoidable. Since hemolysis is An important means of differentiating AIHA from a delayed in onset (typically 3 to 14 days after DHTR relates to the specificity of the antibody(ies) transfusion), the relationship of hemolytic anemia to present in the serum and in a RBC eluate. Some prior transfusion may not be suspected, and a diagnosis antibodies that commonly cause DHTRs have not been of AIHA may seem more appropriate. Further, found or have been reported only rarely as laboratory tests are likely to reveal the presence of a autoantibodies in AIHA. Examples are anti-K and anti- positive DAT and IAT,spherocytosis,and reticulocytosis. Fya, which are frequently encountered in published If multiple alloantibodies or an alloantibody against a cases of DHTRs.6,42–45 Many autoantibodies in warm high-frequency antigen is formed, the findings may be antibody AIHA demonstrate specificity within the Rh difficult to differentiate from AIHA. The diagnostic system but, even here, a distinction between problem is compounded by the fact that, in some cases, autoantibodies and alloantibodies with Rh specificity is AIHA may actually develop as a consequence of blood 1 often possible. Whereas alloantibodies demonstrate transfusion. Indeed, therapy with corticosteroids for truly specific reactions and give clearly negative suspected AIHA has been instituted5,39 or 40,41 reactions with cells lacking the appropriate antigen, contemplated in some reported cases of DHTRs autoantibodies commonly demonstrate “relative before the correct diagnosis was made. specificity.” That is,autoantibodies that are described as having specificity within the Rh system react more Diagnostic Aids strongly or to a higher titer against RBCs bearing a If a patient has been transfused within recent particular Rh antigen, but they will nevertheless react weeks, careful evaluation is needed to distinguish with RBCs lacking the antigen. Thus,a truly specific Rh

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 173 L.D. PETZ antibody strongly suggests that it is an alloantibody, broad range of reactivity, the distinction may be whereas an antibody demonstrating “relative difficult. specificity” is characteristic of autoantibody. A final note of caution is that the IHA may be A further clue to the differentiation of neither a DHTR nor AIHA, but instead may be drug- autoantibodies and alloantibodies having Rh specificity induced IHA. In particular, cephalosporin drugs are is the specificity itself. That is, anti-e is a rare cause of a frequently used in association with surgical procedures DHTR,6 but it is the most frequently described that may require transfusion. In this setting, abrupt autoantibody specificity. In contrast, anti-E is the most onset of a positive DAT and hemolysis may be common Rh alloantibody responsible for DHTRs,6,44 misinterpreted as a DHTR or the sudden onset of AIHA. but it is a relatively unusual autoantibody. Again, if pretransfusion RBCs are still available, it will be References possible to use them to distinguish alloantibody from 1. Petz LD, Garratty G. Immune hemolytic anemias. autoantibody with certainty. 2nd ed. Philadelphia: Churchill Livingstone, 2004. If the antibody shows a defined specificity, the 2. Kaplan HS, Garratty G. Predictive value of direct patient’s RBCs should be tested for the relevant antiglobulin test results. Diagn Med 1985;8:29-33. antigen. If it is an autoantibody, the patient’s RBCs 3. Wolach B, Heddle N, Barr RD, Zipursky A, Pai KR, should possess the antigen. It is not always easy to Blajchman MA. Transient Donath-Landsteiner hae- determine the patient’s phenotype, as DAT+ RBCs are molytic anaemia. Br J Haematol 1981;48:425-34. difficult to phenotype and transfused RBCs may be 4. Heddle NM. Acute paroxysmal cold hemoglobin- present. If transfused RBCs are present, several uria. Transfus Med Rev 1989;3:219-29. methods are available for determining the phenotype 5.Croucher BEE, Crookston MC, Crookston JH. of only the patient’s RBCs: Delayed haemolytic transfusion reactions simu- 1) One method depends on separating out the lating auto-immune haemolytic anaemia. Vox Sang younger RBCs (e.g., reticulocytes are presumed 1967;12:32. to be the patient’s own RBCs).46–50 6. Croucher BEE. Differential diagnosis of delayed 2) Another method, which may be more reliable hemolytic transfusion reaction. In: Bell CA, editor. than the reticulocyte method, utilizes flow A Seminar on Laboratory Management of cytometry.51–53 Hemolysis. Washington, DC: American Association 3) A recent approach is to use DNA typing.54–56 of Blood Banks, 1979:151-60. 7. Worlledge SM. The interpretation of a positive Additional approaches direct antiglobulin test. Br J Haematol 1978;39: If the recipient’s DAT was known to be negative 157-62. prior to transfusion, or if the recipient’s RBCs are still 8. Barry KG, Crosby WH. Auto-immune hemolytic available for the performance of the DAT and it is anemia arrested by removal of an ovarian demonstrated to be negative, this can be valuable teratoma: review of the literature and report of a information. An abrupt change in the DAT from case.Ann Intern Med 1957;47:1002-7. negative to positive is strong evidence that the patient 9. Dawson MA, Talbert W, Yarbro JW. Hemolytic has a HTR rather than AIHA. anemia associated with an ovarian tumor. Am J If it is possible to test the donor units of blood, this Med 1971;50:552-6. procedure may be of significance if an antibody having 10. De Bruyere M, Sokal G, Devoitille JM, Fauchet- specificity that could be that of either an alloantibody Dutrieux MC, De Spa V. Autoimmune haemolytic or an autoantibody is detected, such as, for example, anaemia and ovarian tumour. Br J Haematol 1971; anti-E. If by chance, none of the donor units contained 20:83-94. the E antigen, AIHA or an alloantibody-induced HTR 11. Payne D, Muss HB, Homesley HD, Jobson VW,Baird caused by an undetected antibody must be considered. FG. Autoimmune hemolytic anemia and ovarian Thus, with careful serologic testing, it is possible to dermoid cysts: case report and review of the distinguish a DHTR from AIHA in almost all cases. literature. Cancer 1981;48:721-4. However, if pretransfusion RBCs are not available (as, 12. Shashaty GG, Rath CE, Britt EJ. Autoimmune unfortunately,is usually the case), and if the patient has hemolytic anemia associated with ulcerative an alloantibody or a mixture of alloantibodies with a colitis. Am J Hematol 1977;3:199-208.

174 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 Review: evaluation of immune hemolysis

13.Ramakrishna R, Manoharan A. Auto-immune 26. Kajii E, Miura Y, Ikemoto S. Characterization of haemolytic anaemia in ulcerative colitis. Acta autoantibodies in mixed-type autoimmune hemo- Haematol 1994;91:99-102. lytic anemia. Vox Sang 1991;60:45-52. 14. Arner O, Brohult J, Engstedt L, Karlson R, Sallstrom 27. Shulman IA, Branch DR, Nelson JM,Thompson JC, T. Autoimmune hemolytic anemia in ulcerative Saxena S, Petz LD. Autoimmune hemolytic anemia colitis, cured by colectomy. Report of a case.Acta with both cold and warm autoantibodies. JAMA Med Scand 1971;189:275-7. 1985;253:1746-8. 15. Cunningham-Rundles C. Hematologic compli- 28. Sokol RJ, Hewitt S, Stamps BK. Autoimmune cations of primary immune deficiencies. Blood hemolysis: mixed warm and cold antibody type. Rev 2002;16:61-4. Acta Haematologica 1983;69:266-74. 16. Cunningham-Rundles C, Bodian C. Common vari- 29. Ries CA, Garratty G, Petz LD, Fudenberg HH. able immunodeficiency: clinical and immuno- Paroxysmal cold hemoglobinuria: report of a case logical features of 248 patients. Clin Immunol with an exceptionally high thermal range Donath- 1999;92:34-48. Landsteiner antibody. Blood 1971;38:491-9. 17. Oyefara BI, Kim HC, Danziger RN, Carroll M, Greene JM, Douglas SD. Autoimmune hemolytic 30. Lindgren S, Zimmerman S, Gibbs F,Garratty G. An anemia in chronic mucocutaneous candidiasis. unusual Donath-Landsteiner antibody detectable Clin Diagn Lab Immunol 1994;1:38-43. at 37°C by the antiglobulin test. Transfusion 1985; 18. Cummins L, Spearer W,Stevenson S, et al. Wiskott- 25:142-4. Aldrich syndrome:clinical,immunologic,and path- 31. Nordhagen R. Two cases of paroxysmal cold ologic observations. Am J Dis Child 1959;98:579. hemo-globinuria with a Donath-Landsteiner anti- 19. Pinchas-Hamiel O, Mandel M, Engelberg S, Passwell body reactive by the indirect antiglobulin test JH. Immune hemolytic anemia,thrombocytopenia using anti-IgG. Transfusion 1991;31:190-1. and liver disease in a patient with DiGeorge 32. Dacie JV. The haemolytic anaemias. Part II. Auto- syndrome. Isr J Med Sci 1994;30:530-2. immune haemolytic anaemias. 2nd ed. London: 20.Hsu TC, Rosenfield RE, Burkart P, Wong KY, J. & A Churchill Ltd., 1962. Kochwa S. Instrumented PVP-augmented anti- 33. Sokol RJ, Booker DJ, Stamps R. Erythropoiesis: globulin tests. II. Evaluation of acquired hemolytic paroxysmal cold haemoglobinuria: A clinico- anemia. Vox Sang 1974;26:305-25. pathological study of patients with a positive 21. Garratty G, Petz LD. An evaluation of commercial Donath-Landsteiner test. Hematol 1999;4:137-64. antiglobulin sera with particular reference to their 34. Myhre BA. Fatalities from blood transfusion. JAMA anticomplement properties. Transfusion 1971;11: 1980;244:1333-5. 79-88. 35.Honig CL, Bove JR. Transfusion-associated 22. Von dem Borne AE, Engelfriet CP, Beckers D, Van fatalities: review of Bureau of Biologics reports Der Kort-Henkes G, Van Der Giessen M, Van 1976–1978.Transfusion 1980;20:653-61. Loghem JJ. Autoimmune haemolytic anaemias. II. 36. Sazama K. Reports of 355 transfusion-associated Warm haemolysins—serological and immuno- deaths: 1976 through 1985. Transfusion 1990;30: chemical investigations and 51Cr studies. Clin Exp 583-90. Immunol 1969;4:333-43. 37. Linden JV, Kaplan HS. Transfusion errors: causes 23. Sokol RJ, Hewitt S, Stamps BK. Autoimmune haemolysis: an 18-year study of 865 cases referred and effects. Transfus Med Rev 1994;8:169-83. to a regional transfusion centre. Br Med J (Clin Res 38. Williamson L, Cohen H, Love E, Jones H, Todd A, Ed) 1981;282:2023-7. Soldan K. The Serious Hazards of Transfusion 24. Crookston JH. Hemolytic anemia with IgG and (SHOT) initiative: the UK approach to haemo- IgM autoantibodies and alloantibodies. Arch vigilance. Vox Sang 2000(78 suppl);2:291-5. Intern Med 1975;135:1314. 39. Thomson S, Johnstone M. Delayed haemolytic 25. Freedman J, Lim FC, Musclow E, Fernandes B, transfusion reaction due to anti-Jka and anti-M. Rother I. Autoimmune hemolytic anemia with Can J Med Technol 1972;34:159-61. concurrence of warm and cold red cell autoanti- 40. Holland PV, Wallerstein RO. Delayed hemolytic bodies and a warm hemolysin. Transfusion 1985; transfusion reaction with acute renal failure. JAMA 25:368-72. 1968;204:1007-8.

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 175

L.D. PETZ

41. Rothman IK, Alter HJ, Strewler GJ. Delayed overt 50. Vengelen-Tyler V, Gonzales B. Reticulocyte rich hemolytic transfusion reaction due to anti-U RBCs will give weak reactions with many blood antibody. Transfusion 1976;16:357-60. typing antisera (abstract). Transfusion 1985;25: 42. Pineda AA, Brzica SM Jr., Taswell HF. Hemolytic 476. transfusion reaction. Recent experience in a large 51. Garratty G, Arndt PA. Applications of flow cyto- blood bank. Mayo Clin Proc 1978;53:378-90. fluorometry to red blood cell . 43. Mollison PL, Engelfriet CP,Contreras M, eds. Blood Cytometry 1999;38:259-67. transfusion in clinical medicine. 10th ed., Oxford: 52.Griffin GD, Lippert LE, Dow NS, Berger TA, Blackwell Science Ltd., 1997. Hickman MR,Salata KF. A flow cytometric method 44. Pineda AA,Taswell HF,Brzica SM Jr. Delayed hemo- for phenotyping recipient red cells following lytic transfusion reaction. An immunologic hazard transfusion. Transfusion 1994;34:233-7. of blood transfusion. Transfusion 1978;18:1-7. 53. Wagner F. Identification of recipient Rh pheno- 45. Redman M, Regan F, Contreras M. A prospective type in a chronically transfused child by two- study of the incidence of red cell allo- colour immunofluorescence. Transfus Med 1994; immunisation following transfusion. Vox Sang 4:205-8. 1996;71:216-20. 54. Wenk RE, Chiafari PA. DNA typing of recipient 46. Branch DR, Hian AL, Carlson F, Maslow WC, Petz blood after massive transfusion. Transfusion 1997; LD. Erythrocyte age-fractionation using a Percoll- 37:1108-10. Renografin density gradient: application to 55. Reid ME, Rios M, Powell VI, Charles-Pierre D, autologous red cell antigen determinations in Malavade V. DNA from blood samples can be used recently transfused patients. Am J Clin Pathol to genotype patients who have recently received a 1983;80:453-8. transfusion. Transfusion 2000;40:48-53. 47. Brecher M, ed. Technical manual. 14th ed. 56. Castilho L, Rios M, Pellegrino J, Rodrigues A, Costa Bethesda, MD: American Association of Blood FF. Blood group genotyping for the management Banks, 2002. of patients with “warm” antibody-induced 48. Reid ME, Toy PT. Simplified method for recovery hemolytic anemia (WAIHA) (abstract). Blood of autologous red blood cells from transfused 2001;98:62a. patients. Am J Clin Pathol 1983;79:364-6. 49. Renton PH, Hancock JA. A simple method of Lawrence D. Petz, MD, StemCyte, 400 Rolyn Place, separating erythrocytes of different ages. Vox Sang Arcadia, CA 91007; [email protected]. 1964;9:183.

176 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 Case report: exacerbation of hemolytic anemia requiring multiple incompatible RBC transfusions

A.M. SVENSSON,S.BUSHOR,AND M.K. FUNG

RBC transfusions in a patient with a history of autoimmune alloantibodies in these often multitransfused patients. hemolytic anemia (AIHA) can represent both a laboratory and a These alloantibodies may be very difficult to clinical challenge. The development of high-titer low-avidity antibodies and antibodies to high-frequency antigens may further distinguish from the background of warm autoanti- impair the ability to identify compatible donor RBCs. Not bodies, and in cases of high-titer, low-avidity (HTLA) infrequently, incompatible RBCs must be used and the desire to antibodies or antibodies to high-frequency antigens, increase oxygen carrying capacity conflicts with the desire to avoid exacerbating the autoimmune hemolytic process with RBC the workup may be referred to a reference laboratory transfusions. A 66-year-old Caucasian female with coronary artery with access to rare cells and antisera. disease and a history of refractory AIHA had recently developed It is not possible to define a critical Hb level below anemia and required multiple RBC transfusions. The patient had maintained adequate RBC counts with erythropoietin and which transfusion should be administered during an prednisone therapy for the previous 16 months. With the recent exacerbation of hemolysis. The individual’s capacity to worsening of her hemolytic anemia, she had developed angina that adjust to anemia depends not only on the level of Hb was treated with RBC transfusions in an outpatient setting. However, her angina increased as her RBC counts decreased, measured, but also on the rate of decline in Hb levels leading to hospital admission for further management of her and the capacity for cardiac compensation. In cases hemolytic anemia and angina. She subsequently required multiple with a background of cardiovascular disease, clinical incompatbile RBC transfusions despite increased prednisone therapy and did not improve until after coronary artery stent symptoms and signs of cardiac ischemia guide the placement and high dose IVIG therapy. This case demonstrates the threshold for transfusion on an individual-case basis. usefulness of early patient phenotyping in a case of accelerating The decision to transfuse such a patient thus depends hemolytic anemia to aid in donor RBC selection, the value of communicating with clinicians and the patient regarding the use of on the clinical situation and extensive communication least-incompatible RBCs, and the importance of optimizing the with the clinical staff. patient’s clinical condition to avoid ischemia. In addition, it demonstrates the value of repeated attempts with IVIG treatment despite previous refractoriness to this treatment. Materials and Methods Immunohematology 2004;20:177–183. ABO and Rh grouping, and screening for RBC antibodies, were performed by standard tube Key Words: autoimmune hemolytic anemia, multiple techniques. Similarly,RBC phenotyping was performed incompatible transfusions, IVIG therapy by standard tube techniques with commercially Indications for RBC transfusion of patients with available typing sera. Antibody screening and panel warm autoimmune hemolytic anemia (AIHA) include cells were from multiple sources, including Immucor, the risk for cardiac or cerebral ischemia. Transfusion of Inc., Norcross, Georgia; Gamma Biologicals, Inc., these patients may confer a risk of accelerated Houston,Texas; and Ortho-Clinical Diagnostics,Raritan, hemolysis due to stimulation of the autoimmune New Jersey. All samples were tested with at least 11 process, although this is a controversial notion (see different panel cells plus an autocontrol. DATs were reference 1 for a comprehensive review). However, performed using polyspecific anti-IgG, C3d (Bioclone there is also the risk for intravascular or extravascular Ortho), rabbit anti-IgG (Immucor), and anti-C3b, C3d hemolytic transfusion reactions due to the presence of (Bioclone, Ortho)-specific antisera.

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 177 A.M. SVENSSON ET AL.

Eluates were prepared by a rapid acid elution admission her medications included clonidine, technique (Elu-Kit II, Gamma). Antibody enhancement diltiazem, prednisone at a dose of 15 mg/day, weekly was achieved using a PEG-based method (PEP,GTI,Inc., erythropoietin injections, dietary iron supplemen- Waukesha,WI) or treatment of RBCs with freeze-dried tation, and opioid analgesics for chronic lower back papain (Immucor). PEG autoadsorption was per- and lower extremity pain. There had been no recent formed as previously described.2,3 Alloadsorption of changes in her medication or any infectious prodrome serum was performed using untreated RBCs. Other prior to this most recent exacerbation of her hemolytic methods performed by the American Red Cross anemia. National Reference Laboratory are as described in the case report. Recent history Incompatible units were transfused using the in Three weeks prior to admission to the emergency vivo crossmatch procedure, in which 30 to 50 mL of department, the patient had an office visit for donor blood was slowly transfused into the patient, complaints of and mild angina. She had then the transfusion was temporarily stopped and a previously experienced chest pain when her Hct had sample drawn from the patient and examined for fallen below 30%. At this time, the Hb was 10.6 g/dL visible evidence of hemolysis. In the absence of and the Hct was 32.7%. On the basis of her mild angina hemolysis, the remainder of the donor unit was symptoms, a RBC transfusion in an outpatient setting transfused. A second sample was drawn from the was ordered. The blood bank workup demonstrated patient at the end of the transfusion to look for visible strong microscopic reactivity with all antibody hemolysis. This procedure was repeated with each unit screening cells in the antiglobulin phase (but no transfused. reactivity at the immediate spin phase). The auto- control was negative. Serum reactivity was positive to Case Report a dilution of 1:16 and could not be neutralized with A 66-year-old Caucasian woman, previously plasma.A screen for cold agglutinins was negative. The diagnosed with refractory AIHA, presented to the use of papain-treated RBCs resulted in enhanced emergency department with increasingly severe reactivity to 1+ with all antibody screening cells. No hemolysis and angina on exertion, following about 3 distinct specificity was apparent. An attempt to weeks of increasing transfusion needs on an outpatient remove possible autoreactive antibodies by PEG basis. Her Hct had dropped to 22.5% from 33% 4 days autologous adsorption also generated strong to 2+ prior to admission. The Hb was 7.5 g/dL and the WBC microscopic reactivity with all antibody screening count was 22,630/µL, with 88% neutrophils, 4% cells. The serum also reacted with RBCs negative for monocytes, 5% lymphocytes, 1% basophils, 2% Rg, Ch, Kpb,Jsb, and U. metamyelocytes, and 6 nucleated RBCs/100 WBCs. The The DAT was negative and since the patient had platelet count was 513,000/µL, and the reticulocyte not been transfused for more than 1 year, phenotyping count was > 30%.Total bilirubin was 4.5 mg/dL. was performed. The patient was negative for Cw,Fya,K, S, and P1. Crossmatching with random RBC units Past history showed strong microscopic reactivity. The interpre- The patient was group O, D+ and had refractory tation of these serologic findings was that the AIHA diagnosed 6 years previously that required antibodies detected were most consistent with treatments including splenectomy, cyclophosphamide, nonneutralizable HTLA antibodies, but the possibility rituximab, cyclosporine, and high-dose IVIG. A bone of antibodies to clinically significant, high-frequency marrow biopsy performed at the initial presentation of antigens could not be excluded. Following discussions disease showed no evidence of malignancy. For the with the ordering physician, the patient, and her previous 16 months she had not required blood husband, the patient received the two least- transfusions and antibody screening tests had been incompatible K– units by in vivo crossmatch, without negative for the previous 2 years. Additional medical complications. history included coronary artery disease requiring Before admission to the emergency department, bypass graft surgery more than 10 years previously,two the patient had received transfusions of six least- minor strokes 2 years previously, hypertension, incompatible K–, Fy(a–) units by in vivo crossmatch on hyperlipidemia, and four uneventful pregnancies. On three occasions without complications, but at

178 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

Hemolytic anemia and multiple incompatible RBC transfusions increasingly shorter intervals between transfusions. anti-IgG, and 1+ with monospecific anti-C3d, and a Furthermore, the reactions to antibody screening cells saline control using warm saline 37°C washed RBCs and crossmatches had increased to macroscopic levels was negative. Weak autoagglutination of the patient’s (1+). The DAT and autologous control cells remained RBCs could be removed by warm 37°C saline washing. negative. However, RBCs from the patient were noted The patient’s serum reacted with all RBCs, including to weakly autoagglutinate in the saline control. This autologous cells tested by 37°C albumin, IgG effect could be removed with warm 37°C saline antiglobulin test (AGT), and ficin IgG-AGT. RBCs washing of the cells, suggesting the development of a treated with DTT were still reactive. Furthermore,DTT cold agglutinin. Two allogeneic adsorptions using treatment of the patient’s serum changed reactivity antibody screening cells, both with and without PEG from weak reactivity to 3+. The serum was reactive to enhancement, failed to completely remove reactivity a dilution of 1:256. Five adsorptions with rr,r′r′, or r″r″ that persisted against the cells used in the adsorption RBCs did not remove this reactivity. However,after the procedure. Antibodies against preservatives in the fifth adsorption,the remaining reactivity was uniformly screening cells were ruled out by using multiply- weakly positive against panel cells, suggesting that no washed screening cells. No correlation between the strongly reactive antibodies existed against major RBC strength of reactivity and the age of crossmatched RBC antigens. RBCs with the following phenotypes were units was identified. An extensive review of her medi- found reactive at the albumin-IgG-AGT phase: JMH–, cation list found no drugs reported to induce Di(b–), At(a–), Cs(a–), Yk(a–), Jk(a–b–), I–, PP1Pk, –D–, hemolytic anemia, although the patient’s husband did Rhnull, Cde/Cde, cdE/cdE, Lu(a–b–),Yt(a–), Co(a–), Lan–, report a temporal association with the use of the anti- Vel–, Kp(b–), McLeod, Sc–1, and Jo(a–). Finally, the depressant mirtazapine in the past. However, a search antibody reactivity was assessed for clinical of the literature showed no reported association. significance with the monocyte monolayer assay using group O,c–,E–,P1–,S–,K–,Fy (a–);group O,C–,e–,P1–, Extensive serologic workup S–, K–, Fy (a–); and group O, C–, E–, P1–, S–, K–, Fy (a–) Given the increasing strength of reactivity against RBCs, with and without fresh complement, and using allogeneic cells and the shortened RBC survival, there pooled monocytes from two donors. The reactivity of was a concern that the antibodies detected were the patient’s serum was 21.5 percent to 34 percent against a clinically significant, high-frequency antigen. reactive monocytes, consistent with a clinically A more extensive serologic workup was requested significant antibody (normal range is 0–3%). from the American Red Cross (ARC) Blood Services— On the present admission to the emergency New England Region Reference Laboratory in department, the patient had angina on exertion. There Burlington,Vermont. Briefly, it was concluded that no were no ischemic EKG changes and Troponin I was specificity could be identified, but evidence of weak < 0.15 ng/mL (normal < 0.15,indeterminate 0.15–1.50, complement binding (microscopic) was detected positive > 1.50 ng/mL). The blood bank workup at that using an enhanced DAT (5 minute incubation at 37°C), point showed increased panel cell reactivity of 1 to 2+. which suggested the presence of a warm autoantibody. Furthermore, the DAT was 1+ with broad-spectrum It was further concluded that a nonneutralizable HTLA antiglobulin reagent, 1+ with IgG monospecific serum, antibody was present. RBCs lacking the following high- and 1+ with anti-C3 monospecific serum. However,the frequency antigens were tested and found to be saline control was now strongly positive micro- reactive with the patient’s serum: k, I, Jsb,Kpb,Yta,U,Rg, scopically, suggestive of autoagglutination. With warm Ch, Lan, AnWj, Ge2, Ge3, Jra,Vel, PP1Pk, Coa, Joa, Gya, Hy, 37°C saline washing of the patient’s RBCs, only the Yka,Kna, McCa, McCb,Csa,Lub, and JMH. A new broad spectrum and anti-C3–specific reactions specimen was then sent to the ARC National Reference remained reactive at 1+. An eluate was performed and Laboratory for Blood Group Serology in Philadelphia, found to have nonspecific reactivity at the antiglobulin Pennsylvania, for a more extensive evaluation. phase (weak to 1+ microscopic) for all RBCs tested. Testing performed by the ARC National Reference The autologous cell control at 4°C was also 1+. All Laboratory was similarly unable to identify an antibody crossmatched Cw–, Fy (a–), K–, S– RBC units showed 1+ specificity. The sample received by their laboratory to 2+ reactivity. was a later sample. The DAT was 1+ with polyspecific At this point the risk for ischemic damage to the anti-human globulin, negative with monospecific heart due to severe anemia was weighed against the

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 179

A.M. SVENSSON ET AL. risk of exacerbating the autoimmune process improvement. The patient’s medication at time of underlying the hemolytic anemia with the transfusion transfer included prednisone 60 mg/day, erythro- of incompatible RBCs. Because at this point there were poietin, folic acid, and clopidogrel. no clear clinical signs of a developing myocardial infarction, it was felt that transfusion should be held Discussion off, if possible, pending further testing results. Differentiation between a preexisting warm- However, the following night the patient started to reactive autoantibody and an antibody against a high- complain of moderate chest pain at rest. Troponin I frequency antigen appearing after a transfusion is had increased to 0.29,Hb was 8.5 g/dL,and the Hct was difficult. However, critical results in this patient’s 25.6%. The patient subsequently received two 1+ evaluation were a negative DAT and a negative autolo- crossmatch-incompatible units by in vivo crossmatch gous control. However, with continued transfusion, without complications,following a bolus dose of 60 mg positive results with gradually increasing strength were prednisone. She continued on 60 mg of prednisone seen in these tests. With these findings, warm-reactive per day. On day 3,Troponin I peaked at 0.44. Hb had autoantibodies were considered less likely, but could decreased to 7.7 g/dL, the Hct was 23%, and the not be completely excluded, and more thought was patient’s chest pain persisted at the same level, given to the possibility of a HTLA antibody or an necessitating further transfusions. Only 2+ incom- antibody against a high-frequency antigen. A HTLA patible units could be found and the patient received a antibody was initially favored given the weak reactivity W unit of C –, Fy (a–), K–, S–, P1– RBCs by in vivo that persisted to dilutions of 1:16. However, concern procedure without complications. for a clinically significant antibody, especially against a On day 4, Hb was 9.3 g/dL and Hct was 27.8%. It high-frequency antigen, began to grow as the patient was decided that the patient would undergo cardiac began to require more RBC transfusions at shorter time catheterization and coronary artery stents were placed, intervals, and the reactions obtained with panel cells with immediate relief of symptoms. On day 5, Hb had and crossmatches increased to macroscopic reactivity, decreased to 8.0 g/dL, the Hct was 23.9%, and total considered unusual for a HTLA antibody. Although the bilirubin had increased to 5.8 mg/dL, confirming DAT and autologous cell control were initially negative, increasing hemolytic activity. Reactivity of cross- the patient likely had a chronic immune hemolytic matched phenotype matched units (CW–, Fy a(–), K–, anemia that was previously kept under control with S–, P1–) was generally increased to 1+ to 3+; however, erythropoietin and prednisone therapy. Therefore, the two microscopically reactive units were found. Over confluence of an autoimmune hemolytic process, the following 5 days, the patient continued to receive HTLA antibody, and possible antibody against a high- least-incompatible phenotype-matched units without frequency antigen rendered the decision to transfuse immediate complications or evidence of intravascular crossmatch-incompatible RBCs that much more hemolysis. However, there was an increase in the total difficult. bilirubin (7.7 mg/dL on day 6), an increase in the To aid in the selection of RBC units for transfusion strength of reactivity against crossmatched units, and of patients with unidentifiable specificities, pheno- increasing frequency of transfusion requirements (two typing for common antigens of the Rh, K, Jk, Fy, and RBC units every 2–3 days). Reactivity to crossmatched MNS systems should be performed on the patient’s units continued to increase to up to 4+ on day 7. RBCs at the first opportunity prior to RBC transfusion. Following 10 days of high-dose prednisone In this case, the patient had not been transfused in treatment without discernible improvement, high dose more than a year and presented with a negative DAT. IVIG thereapy was attempted with a one-time dose of While difficult, even in patients who have been 1g/kg body weight. Three days later, Hb had reached transfused over the past 3 months, a phenotype can be 10.6 g/dL, Hct was 31.8%, and total bilirubin was 2.1 obtained based on either molecular4–9 or serologic mg/dL. The patient was no longer in need of techniques.10,11 With the knowledge of which major transfusions and could be transferred to a rehabilitation antigens are lacking in the patient, decisions can then unit, where she continued to maintain Hb values be made in identifying appropriately phenotype- around 11 g/dL and Hct well above 30% (range of matched units that would be least likely to generate a 33%–37%). A bone marrow biopsy was scheduled and hemolytic transfusion reaction and would survive then canceled due to the patient’s dramatic longer in the patient’s circulation.

180 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

Hemolytic anemia and multiple incompatible RBC transfusions

When searching for RBC units that are antigen An alternative option that may be available in the negative, the difficulties increase as multiple antigens future is the use of blood substitutes. Hemoglobin- are required to be simultaneously absent. The need for based blood substitutes are currently in phase III using only partially matched or incompatible units clinical trials for various indications and have been arises and choices must be made as to which antigens used successfully in at least one instance for AIHA.14 to ignore in screening for antigen-negative RBC units to The most effective use of hemoglobin-based blood transfuse. The degree of immunogenicity of each substitutes would be to temporarily stabilize the antigen must then be taken into account, as well as the patient until difficult-to-find or rare donor blood units severity of the possible consequences of transfusing could be obtained or until more aggressive each antigen. Certain combinations of antigen-negative immunosuppressive regimens have had a chance to RBC units are more easily found than others and must take effect. In both instances however, there would be also be taken into account in a search. Furthermore, a prolonged requirement for the use of blood results of alloadsorption studies may indicate the substitutes and the introduction of free hemoglobin absence of any antibodies against major RBC antigens into the patient’s circulation would interfere with the and help in guiding which antigens could be more visual assessment of hemolysis. comfortably ignored. In this patient’s situation, The balance between the risks of inadequate alloadsorptions performed at the reference laboratory oxygenation and subsequent ischemic damage on the failed to completely adsorb all antibodies but reduced one hand, and the risks of transfusing incompatible the reactivity to only microscopic reactivity with all RBCs on the other hand,represents a clinical challenge. cells tested. The inference from this finding was that In situations where there is a critical need to optimize while no statement could be made regarding reactivity oxygen delivery by increasing RBC volume but for against high-frequency antigens, no strongly reacting which no compatible units can be found, transfusion antibodies against major RBC antigens were identified. by in vivo crossmatch is unavoidable. In these However, alloadsorptions would need to be performed instances, such as in this case, close communication at 3 to 4 day intervals to rule out their emergence if the with the clinicians is essential to give adequate input patient continued to be transfused with units positive into the decision-making process. In this context it for antigens that the patient lacked. should be emphasized that the in vivo crossmatch has In the absence of the ability to identify antibody a limited value in predicting survival of the transfused specificities and compatible RBC units by routinely RBCs, since one is only looking for acute gross intra- used serologic tube-based methods, alternative vascular hemolysis during the transfusion.Therefore, a methods exist for determining if an antibody may be negative in vivo crossmatch result only indicates that clinically relevant. The monocyte monolayer assay the likelihood of acute intravascular hemolysis and its quantifies rosetting or phagocytosis of antibody- attendant morbidity and mortality is lessened. sensitized cells by monocytes. In this case it indicated Furthermore, in situations where the in vivo cross- that the antibodies in the patient’s serum had clinical match procedure has been repeated multiple times significance, i.e., would be likely to cause decreased with negative results, continuous awareness of the survival of transfused RBCs. In this particular case, the increased risks of transfusing otherwise incompatible monocyte monolayer assay might have been more RBCs is imperative for both the physicians who order useful if donor cell phenotypes were identified that the transfusions and the nurses who administer them. predicted increased RBC survival. Other reported Finally, it is important to communicate with the methods that have been used include 51Cr labeling of clinicians and the patient the exact nature of “least- RBCs12 and flow cytometry.13 The results of such assays incompatible blood”15 and its increased risks,to address may help evaluate the current immunization status and any concerns regarding the in vivo crossmatch help reveal alloimmunization by comparing the procedure, and to further explain why no compatible survival of autologous RBCs with allogeneic RBCs. blood could be found. However,because an antibody may develop at any time Apart from identifying least-incompatible RBC during a period of repeated transfusions, the results units for transfusion, therapeutic options for patients cannot be used to exclude the existence of clinically with AIHA are generally aimed at reducing the significant alloantibodies in the clinical setting beyond clearance of RBCs and reducing the production of the point of the next transfusion. antibodies. Corticosteroids usually constitute the first

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 181

A.M. SVENSSON ET AL. line of treatment. Splenectomy is used when the References patient is refractory to treatment with corticosteroids. 1. Issitt PD, Anstee DJ. “Warm” antibody-induced Immunosuppressive therapy, such as cyclophos- hemolytic anemia (WAIHA). In: Applied blood phamide and azathioprine and short-term use of group serology. 4th ed. Durham, NC: Scientific cyclosporine, is sometimes successful in inducing Publications, 1999: 939-93. remission in these otherwise refractory patients. The 2. Liew YW, Duncan N. Polyethylene glycol in use of rituximab in AIHA, a monoclonal antibody that autoadsorption of serum for detection of allo- causes specific B-cell depletion by targeting the B-cell antibodies (letter). Transfusion 1995;35:713. 15–18 CD20 antigen, has been reported. For patients who 3. Leger RM, Garratty G. Evaluation of methods for are critically anemic and who are refractory to detecting alloantibodies underlying warm auto- transfusion, plasma exchange may be considered, but it antibodies. Transfusion 1999;39:11-6. is highly inefficient at the removal of IgG antibodies, 4. Reid ME, Rios M, Powell VI,et al. DNA from blood and better suited for patients with hemolysis due to samples can be used to genotype patients who IgM antibodies. 19 have recently received a transfusion. Transfusion The immunomodulatory action of IVIG therapy, 2000;40:48-53. although not well understood, has been used 5. Wenk RE, Chiafari FA. DNA typing of recipient successfully for a myriad of autoimmune disorders.20 blood after massive transfusion. Transfusion 1997; Although unresponsive to this therapy in the past, our 37:1108-10. patient responded promptly to administration of IVIG. 6. Legler TJ, Eber SW, Lakomek M, et al. Application There are substantial differences in IVIG, based on differences in purification and chemical stabilization of RHD and RHCE genotyping for correct blood by manufacturers as well as lot-to-lot variations due to group determination in chronically transfused variation in the donor pool, even with the same patients. Transfusion 1999;39:852-5. manufacturing process. Such differences have been 7. Rozman P, Dov T, Gassner C. Differentiation of noted in the use of IVIG as an immunomodulatory autologous ABO, RHD, RHCE, KEL, JK, and FY therapy in solid organ transplantation21 and may blood group genotypes by analysis of peripheral explain the different responses to IVIG in this patient. blood samples of patients who have recently In conclusion, this case illustrates the management received multiple transfusions. Transfusion 2000; of accelerating AIHA with critical levels of Hb in 40:936-42. association with threatened cardiac ischemia and 8. Kroll H,Carl B,Santoso S,et al.Workshop report on possible alloantibodies. It demonstrates the usefulness the genotyping of blood cell alloantigens. Transfus of performing phenotyping in these patients at first Med 2001;11:211-9. opportunity and the necessity to thoroughly work up 9. Reid ME, Lomas-Francis C. Molecular approaches the antibody reactivity in the presence of a negative to blood group identification. Curr Opin Hematol autologous cell control. Furthermore, it exemplifies 2002;9:152-9. the importance of optimization of clinical conditions, 10. Reid ME,Toy P. Simplified method for recovery of in this case performing cardiac catheterization and autologous red blood cells from transfused stent placement to maximize oxygen delivery to the patients. Am J Clin Pathol 1983;79:364-6. heart. In addition,it shows that IVIG may be useful and 11. Branch DR, Hian AL, Carlson F, et al. Erythrocyte should be attempted in these patients even if prior age-fractionation using a Percoll-Renografin treatments with IVIG have been unsuccessful. density gradient:application to autologous red cell antigen determinations in recently transfused Acknowledgments patients. Am J Clin Pathol 1983;80:453-8. We thank the laboratory staff of the Fletcher Allen 12. Baldwin ML, Barrasso C, Ness PM, Garratty G. A Health Care Blood Bank; American Red Cross (ARC) clinically significant erythrocyte antibody detect- National Reference Laboratory for Blood Group able only by 51Cr survival studies. Transfusion Serology in Philadelphia, Pennsylvania; and the ARC 1983;23:40-4. Blood Services-New England Region Reference 13. Zeiler T,Müller JT,Hasse C, et al. Flow cytometric Laboratory in Burlington,Vermont, for their invaluable determination of RBC survival in autoimmune assistance. hemolytic anemia. Transfusion 2001;41:493-8.

182 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

Hemolytic anemia and multiple incompatible RBC transfusions

14. Mullon J, Giacoppe G, Clagett C, et al. Transfusions 19. Silberstein LE, Berkman EM. Plasma exchange in of polymerized bovine hemoglobin in a patient autoimmune hemolytic anemia (AIHA). J Clin with severe autoimmune hemolytic anemia. N Apheresis 1983;1:238-42. Engl J Med 2000;342:1638-43. 20. Knezevic-Maramica I, Kruskall MS. Intravenous 15. Petz LD. Least incompatible units for transfusion immune globulins: an update for clinicians. in autoimmune hemolytic anemia: should we Transfusion 2003;43:1460-80. eliminate this meaningless term? A commentary 21.Wassmuth R, Hauser IA, Schuler K, et al. for clinicians and transfusion medicine Differential inhibitory effects of intravenous professionals.Transfusion 2003;43:1503-7. immunoglobulin preparations on HLA-alloanti- 16.Ahrens N, Kingreen D, Seltsam A, Salama A. bodies in vitro. Transplantation 2001;71;1436-42. Treatment of refractory autoimmune haemolytic anaemia with anti-CD20 (rituximab). Br J Annika M. Svensson, MD, PhD, Sharon Bushor, Haematol 2001;114:244-5. MT(ASCP), and Mark K. Fung, MD, PhD (correspon- 17. Quartier P,Brethon B, Philippet P,et al. Treatment ding author), Fletcher Allen Health Care/University of childhood autoimmune haemolytic anaemia of Vermont Department of , 111 Colchester with rituximab. Lancet 2001;358:1511-3. Avenue, Burlington, VT 05401. 18. Morselli M, Luppi M, Potenza L, et al. Mixed warm and cold autoimmune hemolytic anemia: complete recovery after 2 courses of rituximab treatment. Blood 2002;99:3478-9.

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 183

Delayed hemolytic transfusion reaction due to anti-Fyb caused by a primary immune response: a case study and a review of the literature

H.H. KIM,T.S.PARK, S.H. OH, C.L. CHANG,E.Y.LEE,AND H.C. SON

Delayed hemolytic transfusion reactions (DHTRs) usually occur Case Report between 3 and 14 days posttransfusion as a result of a secondary immune response, with a drop in Hb level, fever, jaundice, or A 42-year-old man suffering from multiple fractures hemoglobinuria. DHTRs caused by a primary immune response are was admitted to the emergency room of Pusan National particularly rare events, and only a few reports have been known. University Hospital on January 15, 2003, after falling In this report, we describe an unusual case of a DHTR caused by while he was on duty at a construction site. He was anti-Fyb in a 42-year-old man, who had no prior history of transfusion. Although it seems to be a rare phenomenon, we scheduled for emergency surgery since an L-Spine MRI suggest that DHTRs by a primary immune response may be revealed an unstable bursting fracture (L1) with cord considered even in the case of the patient who had typical compression. Right distal radius fracture and right evidence of hemolysis but who had no previous transfusion history. Immunohematology 2004;20:184–186. calcaneal fracture were also present. His RBCs typed as group O,D+. Antibody screening was negative. During Key Words: delayed hemolytic transfusion reactions, surgery, the patient received three units of packed anti-Fyb, primary immune response RBCs and two units of FFP. Postsurgical bleeding caused the patient’s Hb level to continuously fall. Over The most important Duffy antigens in routine the following 7 days, he received an additional 18 units blood bank serology are Fya and Fyb.1 Because Duffy of saline crossmatch-compatible packed RBCs until the antigens are only moderate immunogens, anti-Fya Hb level stabilized at 11.5g/dL. The prothrombin time occurs with one-third the frequency of anti-K. In was mildly increased to 18.7 seconds (reference range addition, anti-Fyb antibodies have an incidence rate of 1 11–14.1). On the 11th hospital day, jaundice and an to 20 compared to anti-Fya.2 However, these alloanti- unexpected fall in Hb (9.9 g/dL) occurred. Antibody bodies are usually IgG; react best at the antiglobulin screening was performed again. Anti-Fyb was identified phase; and are known as clinically significant, and confirmed by gel test using LISS/Coombs and unexpected antibodies, related to acute hemolytic NaCl/Enzyme cards (DiaMed AG, Cressier Sur Morat, transfusion reactions (HTRs) and delayed hemolytic Switzerland). The patient’s RBCs were negative in the transfusion reactions (DHTRs).1,3 IgM Duffy antibodies DAT. The patient had no previous history of transfusion usually occur as a primary immune response. IgG nor history of any particular medical or surgical Duffy antibodies form via a secondary immune illnesses, except that he had received minor surgery on response after another exposure to Duffy antigens.4,5 one of his right toes 5 years previously. The laboratory DHTRs usually become apparent between 3 and 14 findings on the 11th hospital day were as follows: total days posttransfusion and are generally attributed to an bilirubin 2.15 mg/dL (reference range 0.3–1.3 mg/dL), anamnestic immune response. HTRs caused by a direct bilirubin 0.87 mg/dL (reference range 0.05–0.40 primary immune response are extremely rare events, mg/dL), LDH 1146 IU/L (reference range 218–472 and only a few reports have been published.6–9 IU/L), C3 121 mg/dL (reference range 50–90 mg/dL),

184 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

Primary response: anti-Fyb and delayed transfusion reaction

C4 23.5 mg/dL (reference range 10–40 mg/dL), and C- primary immune response was reported by Patten et reactive protein (CRP) 5.17 mg/dL (reference range al.7 Solanki et al.8 described three patients with no 0–0.5 mg/dL). The haptoglobin was below detectable transfusion history who had DHTRs caused by a levels (< 10.0 mg/dL), but the level of alpha-1-acid primary immune response. Recently, a DHTR due to a glycoprotein was 171.0 mg/dL (reference range 45–98 primary immune response that stimulated anti-Jka and mg/dL). The patient was diagnosed with a DHTR -K in a 24-week-old female was reported.9 However,no caused by anti-Fyb. Three compatible units of Fy(b–) known report has yet been published on a DHTR by a packed RBCs were issued after crossmatching using primary immune response involving Fyb, an antigen the antiglobulin method with LISS/Coombs card which has relatively low immunogenecity. (DiaMed AG). No further hemolytic reactions oc- Haptoglobin depletion is usually the most sensitive curred, and the unexpected antibody, present in his laboratory indicator of hemolysis. On the other hand, serum for 46 days after antibody detection, could no haptoglobin synthesis is increased in the presence of longer be detected. acute inflammatory processes. In our patient, we observed an undetectable haptoglobin level but high Materials and Methods alpha-1-acid glycoprotein and CRP,so the possibility of extravascular hemolysis plus an acute phase response For detection of RBC antibodies, column must be considered. Unfortunately, we could not test agglutination methods were used. The DAT was the urine or test for plasma Hb because of the presence performed on the patient’s RBCs from an EDTA- of hematuria caused by trauma and because a plasma anticoagulated sample using the polyspecific LISS/ specimen was not available. Coombs card (DiaMed AG). Duffy phenotypes were determined on RBCs from EDTA-anticoagulated samples using ID-Antigen profile III (DiaMed AG). The Conclusion RBCs used as screening and identification panels were On the basis of clinical and laboratory findings, as from commercially prepared reagent panel cells well as the patient’s past history, DHTR by a primary (DiaMed AG). Antibody screening and identification immune response was strongly suspected,even though tests were performed with LISS/Coombs and NaCl/ DHTRs caused by a primary immune response are Enzyme cards (DiaMed AG) at 37°C for 15 minutes. extraordinarily rare events. It is thought that such an DTT (Sigma Chemical Co.,St.Louis,MO) treatment was atypical hemolytic reaction must be related to the also performed to determine the immunoglobulin class transfusion of the large amount of packed RBCs within of the alloantibody in the patient’s serum. a short period of time. Thus, although it is a very rare phenomenon, we suggest that DHTR by a primary Results immune response should be considered, even in the case of the patient who had typical evidence of The serum of the patient was reactive with all hemolysis but no previous transfusion history. Fy(b+) RBCs and negative with all Fy(b–) RBCs. Anti- Fyb was not detectable in the enzyme phase. The Acknowledgment patient’s RBCs were Fy(a+b–) and were negative in the We thank Dr. S. Gerald Sandler for many insightful DAT when anti-Fyb was detected. DTT treatment of the comments and his recommendation, and MIRR SciTech patient’s serum did not alter reactions, suggesting that Corp. for technical assistance. the anti-Fyb was an IgG alloantibody. References Discussion 1. Harmening DM. Modern blood banking and trans- A study of 11 cases related to DHTRs by a primary fusion practices. 4th ed. Maryland: F.A.Davis,1999: immune response in burned children, who had never 161-99. been transfused, was reported by Bacon and co- 2. Marsh WL, Ehrich CC. The Duffy blood group workers.6 All 11 patients had a negative DAT and IAT system: a review of recent developments. before transfusion. In this study,anti-K and anti-E were Infusionsther Klin Ernahr 1975;2:280-9. the most frequently identified antibodies. The authors 3. Brecher ME. Technical manual. 14th ed. Bethesda, also reported DHTRs due to anti-S, -C, and -Jka, MD: American Association of Blood Banks, 2002: respectively. Another case of DHTR due to anti-C by a 315-59.

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 185

H.H. KIM ET AL.

4. Mollison PL, Engelfriet CP, Contreras M. Blood 8.Solanki D, McCurdy PR. Delayed hemolytic transfusion in clinical medicine. 10th ed. Oxford: transfusion reactions. An often-missed entity. Blackwell Science, 1997:81-2. JAMA 1978;239:729-31. 5.Anderson KC, Ness PM. Scientific basis of 9. Albiero AL, Novaretti MC, Llacer PE, Chamone DA. transfusion medicine. 1st ed. Philadelphia: Early primary immune response against erythro- Saunders, 1994:507-8. cytes: a case report. Transfus Med 2003;13:93-7. 6. Bacon N, Patten E, Vincent J. Primary immune response to blood group antigens in burned Hyung Hoi Kim, MD, PhD (corresponding author), children. Immunohematology 1991;7:8-11. Tae Sung Park, MD, Seung Hwan Oh, MD, Chulhun L. 7. Patten E, Reddi CR, Riglin H, Edwards J. Delayed Chang, MD, PhD, Eun Yup Lee, MD, PhD, and Han hemolytic transfusion reaction caused by a Chul Son, MD, PhD, Department of Laboratory primary immune response. Transfusion 1982;22: Medicine, College of Medicine, Pusan National 248-50. University, 1-10 Ami-dong, Seo-gu, Busan 602-739, Korea.

186 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

Maternal alloanti-hrS—an absence of HDN

R. KAKAIYA,J.CSERI,B.JOCHUM,L.GILLARD,AND S. SILBERMAN

A 24-year old female, gravida III, para III, delivered a full-term infant of the antibody response in individuals whose RBCs by cesarean section. A maternal blood sample at the time of express a variant e antigen or lack some epitopes of the admission showed antibody in her serum that had apparent anti-e specificity and that her RBCs were e+. Further studies determined e antigen. that the antibody was anti-hrS. Cord RBCs had a negative DAT and a normal Hb level. There was no clinical evidence for increased Case Report hemolysis in the infant. We describe an hrS+ infant with no evidence of HDN due to anti-hrS. Immunohematology A 24-year-old African American female, gravida III, 2004;20:187–189. para III, presented for elective cesarean section in October 2003. Maternal history included two previous Key Words: anti-hrS, variant e antigen, HDN due to cesarean sections with the last delivery in February anti-hrS 2000, at which time an anti-e was detected in the The e antigen is present in about 98 percent of the maternal serum. The second child was not affected Caucasian population. The hrS antigen, a variant form with HDN. No information was available for the first of the e antigen, is a high-frequency Rh antigen present pregnancy and delivery. The father’s ethnic origin is on all cells except for e– RBCs and rare e+ RBCs that unknown and he was not available for testing. lack hrS. Alloanti-e–like antibodies may be made by those individuals with e+ RBCs lacking this variant Results S antigen. The hr antigen was first reported in 1960 by Maternal sample Shapiro, in a Bantu patient whose serum reacted with A maternal blood sample obtained before the 1 all cells bearing E or e. Shapiro estimated that about 6 cesarean section was tested during the present percent of Bantu Rh haplotypes were Dce or dce admission. The RBCs were A+, C–, E+, c+, e+. A DAT S encoding no hr antigen. Following adsorption of the was negative. Serum tested in a routine LISS panel patient’s serum with R2R2 RBCs, the serum no longer (Immucor-Gamma, Houston, TX) showed no reactions reacted with E+e– RBCs but still reacted with most e+ at immediate spin,a weak reaction with eight of eleven 2 RBCs. The antigen, defined by the antibody reacting cells at 37°C, and micro to weak reactions with seven only with e+ RBCs (except for a rare e antigen detected of eleven cells at the antihuman globulin (AHG) phase. in black South Africans) was named hrS and the broader A routine PEG panel (Immucor-Gamma) revealed specificity reacting with E+e+ RBCs was named anti-Hr apparent anti-e specificity with 1+ to 2+ reactions at (later, anti-Rh18). Thus, anti-hrS should be suspected in the AHG phase. An autocontrol run with both panels patients with apparent alloanti-e specificity whose was negative. Serum was also tested against a seven- RBCs are positive for the e antigen. This antibody has member select-cell PEG panel consisting of e– RBCs. been described as analogous to the anti-D made by D+ The e– PEG panel was negative. individuals. Because the patient’s RBCs were e+, the possibility Some anecdotal reports suggest that antibodies to that the maternal antibody might be anti-hrS and not portions of the e antigen may be clinically significant.2 anti-e was considered. To confirm this possibility, the According to Reid and Lomas-Francis,3 the available serum was tested against two RBC samples that were data for the clinical significance of this alloantibody in e+, hrS–. This testing showed negative reactions with HDN are limited. We describe an hrS+ infant with no both cells. In addition, positive reactions were seen evidence of HDN due to anti-hrS and review some of with the maternal serum against two RBC samples that the available information concerning the heterogeneity were e+ and hrB–, ruling out the presence of anti-hrB in

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 187

R. KAKAIYA ET AL. the maternal serum. At the time of these tests, an anti- transfusions of incompatible RBCs. Therefore, hrS was not available to confirm the patient’s RBCs as transfusion in patients with these antibodies should be hrS–. The titer of the maternal serum versus rr RBCs approached with caution. According to Moores, the without the addition of enhancement media was as immune response of hrS– people is highly variable.5 follows: undiluted,2+;1:2,1+;1:4 and 1:8,microscopic The results of the cord blood DAT in her report varied reactions. from microscopic positive to 4+ due to anti-Rh18. Seven affected infants received successful exchange Child’s sample transfusions, although one died later. In addition, three A cesarean section was performed and a healthy infants suffered mild hemolytic disease, and seven baby girl was delivered. The cord blood was O+ with a other infants either were miscarried or were delivered negative DAT. Antibody screen on the cord serum was elsewhere. Grobbelaar and Moores6 described an anti- negative. At birth, the child’s Hb level was 18.8 gm/dL hrS which would now be considered an anti-Rh18 in and the Hct was 56.5%. The total bilirubin levels on the the serum of a Bantu woman whose newborn suffered first day of life ranged between 2.5 and 4.0 mg/dL. from mild HDN. In our case,the maternal antibody was Clinically and serologically,the baby did not show signs anti-hrS (not anti-Rh18) and was not clinically of hemolysis; she was discharged on the third day. significant. This was our patient’s third pregnancy and At 2 months of age, the child’s blood sample was at least the last two of her infants were unaffected. The obtained and tested. The child tested positive for e and antibody tested only weakly with LISS and was stronger hrS antigens. (At this time an anti-hrS from a patient was with PEG. Moreover, this antibody did not react with available to test the child’s RBCs). A complete Rh RBCs that are both E+ and e+, the child’s phenotype. phenotype was determined with the following results: The reasons for the absence of HDN in our case are D+, C–, c+, E+, e+, and hrS+. unclear but could be related to the nature or strength of maternal alloantibody and/or a decreased antigen Discussion density on fetal RBCs. Noizat-Pirenne et al.7 determined that several In our case, the maternal antibody in the serum S appeared to have anti-e specificity on routine testing genetic events give rise to the absence of hr in Black using LISS and PEG panels. The reactions were weak individuals. Specifically, she determined that loss of with a LISS panel, but stronger with a PEG panel. The certain amino acid residues in a transmembrane maternal RBCs were e+. Because an apparent alloanti- domain might alter the protein configuration and the e was present in the serum of a patient who was e+,we immune response to the antigen. suspected the possibility that the patient may have an The ability to identify alleles responsible for alloantibody to the e-variant hrS. Therefore, we under- variable expression of the e antigen likely will have took further testing using RBCs that were e+, hrS–. practical application. For instance, fetal DNA obtained Using these rare RBCs, we were able to confirm the from maternal plasma during pregnancy can be presence of anti-hrS in the maternal serum, suggesting examined to determine if the allele of interest was that the patient’s RBCs were hrS–, a variant form of the transmitted to the fetus. e antigen. Testing of the infant revealed an absence of References serologic or clinical signs of hemolysis, including a 1. Shapiro M. Serology and genetics of a new blood negative DAT on the cord blood sample. At 2 months factor: hrS. J Forensic Med 1960;7:96-7. of age,the child’s RBCs were tested and were shown to 2. Issitt PD, Anstee DJ. The antigen hrS. In: Applied be e+ hrS+. These findings show that the maternal anti- blood group serology. 4th ed. Durham, NC: hrS failed to cause HDN. Montgomery Scientific Publications, 1998:368-72. Unpublished observations suggest that some 3. Reid ME, Lomas-Francis C. hrS antigen. In: The antibodies produced in people with a e-variant blood group antigen factsbook. New York: phenotype can be significant in vivo and sometimes Academic Press, 1997:119-20. effect clearance of small labeled aliquots of 4. Noizat-Pirenne F,Lee K, Le Pennec PY, et al. Rare incompatible RBCs. However, recent data from Noizat- RHCE phenotypes in black individuals of Afro- Pirenne et al.4 show that anti-Rh18 (containing anti-hrS) Caribbean origin: identification and transfusion caused fatal transfusion reactions in two patients after safety. Blood 2002;100:4223-31.

188 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

Alloanti-hrS—absence of HDN

5.Moores P. Rh18 and hrS blood groups and Ram Kakaiya, MD, Medical Director, LifeSource antibodies. Vox Sang 1994;66:225-30. Blood Services, 1205 North Milwaukee Avenue, 6. Grobbelaar BG, Moores P. The third example of Glenview, IL 60025; Jill Cseri, MT(ASCP)SBB, anti-hrS. Transfusion 1963;3:103-4. LifeSource Blood Services, Beth Jochum, 7. Noizat-Pirenne F,Mouro I, Le Pennec PY,et al. Two MT(ASCP)SBB, LifeSource Blood Services, and Laurie new alleles of the RHCE gene in Black individuals: Gillard, MT(ASCP)SBB, and Simone Silberman, MD, the Rhce allele ceMO and the RhcE allele cEMI.Br Loyola University Medical Center, Maywood, IL. J Haematol 2001;113:672-9.

Attention: State Blood Bank Meeting Organizers If you are planning a state meeting and would like copies of Immunohematology for distribution, please contact Mary McGinniss, Managing Editor, 4 months in advance, by phone or fax at (301) 299-7443.

Attention SBB and BB Students: You are eligible for a free 1-year subscription to Immunohematology. Ask your education supervisor to submit the name and complete address for each student and the inclusive dates of the training period to Immunohematology, P.O. Box 40325, Philadelphia, PA 19106.

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 189

COMMUNICATIONS

Letter to the Editors

HAMA (Human Anti-Mouse Antibodies) do not Cause False Positive Results in PAKPLUS

Regretfully,we wish to inform readers that a recent glycoproteins are affinity purified and immobil- article which appeared in Immunohematology (False ized directly. Therefore, the suggestion that a reactivity in GTI PAKPLUS ELISA kits due to the HAMA can cause false positive results in the presence of anti-mouse antibody in patient’s samples, GPIb/IX, GPIV, and HLA wells is untenable. MF Leach, JP AuBuchon, Volume 19, No.4, p 112.) (3) Lastly,false positive results due to HAMA are very contained several significant errors. unlikely because the specimen diluent provided In Figures 1 and 2 the authors provide illustrations with the GTI PAKPLUS kit already contains of how a human anti-mouse antibody (HAMA) can mouse immunoglobulin. The amount of cause both false positive and false negative results in immunoglobulin present in the diluent was testing done with the GTI kits. On the one hand, they optimized to prevent interference from HAMA. claim that a false positive result can be caused by The use of this diluent in PAKPLUS has been binding of a HAMA to the mouse monoclonal antibody tested using well-characterized samples con- used to immobilize platelet antigens in the well. On taining HAMA. None was found to give a the other hand, they suggest that a false negative result positive result in the PAKPLUS kit. This includes can be caused by binding of a HAMA to the secondary HAMA induced by treatment with therapeutic antibody-enzyme conjugate, thereby preventing it from reagents containing mouse monoclonal anti- recognizing a patient antibody bound to the target bodies and those naturally occurring in human antigen (resulting in “neutralization” of the secondary populations. antibody-enzyme conjugate). We would also like to add that, although We wish to point out that these suggestions are commercial ELISA assays are available for confirming flawed for the following reasons: the presence of HAMA antibodies, none of the samples (1) The PAKPLUS kit is not a simultaneous sandwich in this study were tested by the authors to confirm the immunoassay (i.e., the sample is not simul- HAMA specificity. taneously incubated with both the capture and The authors recommended that mouse IgG should the detection antibody reagents). As stated in the be added routinely to all patient samples prior to PAKPLUS direction insert, there is a wash step testing. We strongly disagree with them on this between the addition of the patient sample and point. Users are generally not aware of all of the the addition of the secondary antibody-enzyme complexities that underlie a commercial assay. conjugate. Thus, any unbound antibody, (includ- Manipulating the components without a full under- ing HAMA) would be removed from the test well standing of these complexities can easily lead to prior to the addition of the secondary antibody- erroneous results. Finally, the mouse IgG used by the enzyme conjugate. Therefore, a false negative authors was not knowingly provided by GTI for the result due to HAMA antibodies could not occur type of study done by the authors. in the way described by the authors in Figure 2. Leigh Ann Tidey, MS, MT(ASCP)SBB (2) The authors claim to have identified HAMA antibodies that cause false positive results with Director of Operations each of the five glycoproteins immobilized in the QA Manager wells. However, as described in the package GTI insert, only two glycoproteins in PAKPLUS are Suzette Chance, PhD captured in the wells by the use of immobilized Director of Product Development monoclonal antibodies. The other three GTI

190 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

COMMUNICATIONS CONT’D

Marilyn Clarke, PhD to the problem we encountered. We thank GTI for Production/Process Scientist suggesting this approach to us. GTI The presence of mouse immunoglobulin in the RH Aster, MD specimen diluent of this kit as routinely supplied by Senior Investigator the manufacturer undoubtedly helps reduce the Blood Research Institute frequency of this problem. This amount apparently The Blood Center of Southeast Wisconsin may not be sufficient to neutralize anti-mouse Milwaukee WI reactivity in some samples. We attempted to identify results that might make the user of the test kit The above letter was sent to Leach and AuBuchon. suspicious for the unexpected effect of the heterophile They offered the following reply. antibodies that eluded neutralization in order to prompt the use of additional mouse immunoglobulin 1 We appreciate the interest of Tidey et al. in our to obtain an accurate result. As this and many other recent studies of the effect of heterophile antibodies in laboratories depend on these test results to detect and 2 enzyme immunoassays for platelet alloantibodies. We characterize alloantibodies in order to provide were frankly surprised at many of their comments effective platelet hemotherapy, we hope that others inasmuch as it was a staffmember of their company may find this supplemental technique useful in certain that first led us to consider the presence of this situations. phenomenon as an explanation for some anomalous Researchers in our laboratory have enjoyed results we had experienced and provided (on June 9, successful collaborations with many manufacturers in 2000) mouse immunoglobulin and instructions for its developing and validating a wide variety of techniques use to allow us to investigate this problem. and equipment. We have found open communications The authors of the letter contend that the washing essential in these efforts to improve our collaborative step before the addition of the conjugate precludes a service to patients, and our communications to GTI heterophile antibody from remaining in the well. in investigating the situation we were encountering However, the capability of heterophile antibodies to were no different. The letter’s implication of nefarious bind with sufficient avidity such that they remain in conduct is unwarranted and unsubstantiated. We the test system to be recognized by the secondary appreciate GTI’s provision of mouse IgG to investigate antibody-antigen conjugate has been seen in many this problem after their staff had alerted us to its other test systems. Thus, when we encountered possibility,and we offered GTI review of an abstract of unexplained reactivity in samples that were not the work3 (on April 9, 2002) at the same time that we expected to contain platelet alloantibodies, and when requested additional mouse immunoglobulin to GTI’s suggestion to add (additional) mouse immuno- continue our investigations. We regret that the globulin removed this reactivity, we felt it was not manufacturer did not pursue this opportunity. unreasonable to conclude that heterophile antibodies Enzyme immunoassays remain an important tool had been the cause. The authors of the letter also for the detection of platelet alloantibodies. As stated that the antigens in some wells are immobilized additional labs report their experiences with using such that an anti-mouse antibody would have no target. these techniques in “real life” situations, further While we are not privy to the details of the manu- knowledge may accrue that facilitates improvement of facture of the kit and did not directly analyze samples their useful attributes. for the presence of anti-mouse antibodies, our observation that the addition of murine immuno- globulin blocks otherwise inexplicable activity is prima facie evidence of the presence of anti-mouse antibody reactivity and an obvious and simple solution

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 191

COMMUNICATIONS CONT’D

References Miriam Fogg Leach, MS, MT(ASCP)SBB 1. Tidey LA, Chance S, Clarke M, Aster RH. HAMA James P. AuBuchon, MD (human anti-mouse antibodies) do not cause false Department of Pathology positive results in PAKPLUS. Immunohematology Dartmouth-Hitchcock Medical Center 2004;20: One Medical Center Drive 2. False reactivity in GTI Pak Plus® ELISA kits due to Lebanon, NH 03756 the presence of anti-mouse antibody in patients’ samples. MF Leach, JP AuBuchon. Immuno- hematology 2003;19:112-7. 3. Leach MF, AuBuchon JP. False positive results in GTI-Pak Plus ELISA kits due to the presence of anti-mouse antibodies in patient samples. Transfusion 2002;42:64S.

Notice to Readers: Immunohematology, Journal of Blood Group Serology and Education, is printed on acid-free paper.

192 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

COMMUNICATIONS CONT’D

Letter to the Editor-in-Chief

Immunohematology to be listed in Index Medicus and MEDLINE

S. Gerald Sandler, MD

In February 2004, I was informed by the National (3) I provided an article-by-article critique of the three Library of Medicine (NLM) that its Literature Section most recent issues of Immunohematology, Technical Review Committee had completed a explaining to the reviewers (I assumed none were favorable review of Immunohematology and that the “blood bankers”) what each article contributed to NLM would begin to cite and index the journal in our discipline. I identified each of the authors and, Index Medicus and MEDLINE. The NLM’s decision is a I must admit, the list of contributors was truly key milestone in the history of Immunohematology.It distinguished and impressive. means that articles in Immunohematology—past, Compiling this information to illustrate Immuno- present, and future—will be listed on electronic data- hematology’s high standards and quality was both an bases, making them readily accessible to researchers, authors, and readers worldwide. Preparing the appli- inspiring and a sentimental experience. I recall, as cation for the NLM’s review of Immunohematology though it were only yesterday, that Delores Mallory was an informative and inspiring experience. I would arrived at the National Headquarters of the American like to share some of my observations of that Red Cross to direct our newly relocated National experience with you. Reference Laboratories in Bethesda, MD. Her The NLM’s Technical Review Committee had responsibilities included coordination of the National declined three prior applications for Immuno- Reference Laboratory Committee, which published the hematology to be listed in Index Medicus and Red Cell Free Press. Delores had a vision for the Red MEDLINE. In 2003, I approached Delores Mallory, Cell Free Press. She believed that there was a need and Editor-in-Chief, and volunteered to initiate a fourth an opportunity to dress up the newsletter and establish attempt. I was convinced that Immunohematology, as a quarterly journal to be named Immunohematology, well as its distinguished contributors, deserved this Journal of Blood Group Serology and Education. In recognition and status. I drafted an extensively refer- 1986, she recruited Mary McGinniss, who had recently enced appeal to the NLM’s Technical Review retired from a distinguished career at the National Committee, basing arguments on the following three Institutes of Health, to assist in scientific editing. observations: Within a few years, Mary was appointed Managing (1) Immunohematology serves a unique niche among Editor of Immunohematology. Volume 1, number 1 of scientific publications. It is the only journal Immunohematology appeared in September 1984. worldwide that limits its focus to blood group Delores announced the change and her vision in a page serology. No other journal, including Transfusion, 1 editorial, as follows: Blood, or Vox Sanguinis, targets a readership of “The title of our newsletter—Red Cell Free Press— blood group serologists and immunohematologists has been put to rest and in its place is a new name— to provide up-to-date practical information. Immunohematology—and a new direction. There (2) Immunohematology’s editors and editorial board will be more articles relevant to blood group are the international leaders in blood group serology and education . . .” serology. I listed the members of the editorial board and, one by one, cited examples of each Contributors to this first issue included Rebecca H. member’s scientific contributions and expertise in Buckley, MD; Herbert A. Perkins, MD; Peter Issitt, PhD; the field of blood group immunohematology. Kay Beattie, MT(ASCP)SBB;Tabbie Bolk, MT(ASCP)SBB;

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 193

COMMUNICATIONS CONT’D

Judy Robinson; Mary N. Crawford, MD; Denise A.Valko, had an opportunity to share in your progress are proud MS, MT(ASCP)SBB; Joan Barker, MT(ASCP)SBB; Roger of your achievements and progress. Collins; and Dorothy C. Malamut, SBB(ASCP). The S. Gerald Sandler, MD journal’s standards were set high from the beginning Associate Medical Editor, Immunohematology and these high standards have been maintained for 80 Professor of Medicine and Pathology quarterly issues during the past 20 years. Happy 20th Georgetown University Medical Center birthday, Immunohematology! Those of us who have Washington, DC

IMPORTANT NOTICE ABOUT MANUSCRIPTS FOR IMMUNOHEMATOLOGY Mail all manuscripts (original and 2 copies) to Mary H. McGinniss, Managing Editor, 10262 Arizona Circle, Bethesda, MD 20817. In addition, please e-mail a copy to Marge Manigly at [email protected]

194 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

COMMUNICATIONS CONT’D

Letters From the Editor-in-Chief

Ortho dedication

14 years! Thank you Ortho-Clinical Diagnostics for Please let your Ortho-Clinical Diagnostics the financial support needed to publish the September representatives know how much you appreciate their issue of Immunohematology for the 14th year in a support of Immunohematology. It means a great deal row! It demonstrates once again the commitment to the publication of this journal! Thank you! Ortho-Clinical Diagnostics has to the support of education and to the advancment of scientific knowledge and original ideas.

The final 20th anniversary issue

The final 20th anniversary issue is a grand finale was the Rare Donor Registry of the American Red that you should not miss! Ralph R.Vassallo, MD, Senior Cross. Dr Greenwald started both programs. The two Medical Director of the American Red Cross, Penn- programs were combined into the American Rare Jersey Region,is the final guest editor and he has a four- Donor Program which is now housed in the Penn- star, two-thumbs-up issue to finish the year. There will Jersey American Red Cross. One of the invited review be five invited reviews: The Function of Blood Groups articles in the December issue is a report of the by Jill Storry,PhD, FIBMS; Immunohematologic Aspects American Rare Donor Program by Ann Church and co- of Transplantation by Jeffrey McCullough, MD, et al.; authors. IgA Anaphylactic Transfusion Reactions: I. Laboratory Dr Greenwald was honored this year by the Diagnosis, Incidence, and Supply of IgA-Deficient American Association of Blood Banks with the Karl Products by Ralph R.Vassallo, MD; IgA Anaphylactic Landsteiner Memorial Award, the highest award given Transfusion Reactions: II. Clinical Diagnosis and by the association. Congratulations, Dr. Greenwald. Management by S. Gerald Sandler, MD; and The Well done and thank you for your contributions to American Rare Donor Program by Ann Church,Cynthia transfusion medicine and the American Rare Donor Flickinger, and Tammy Petrone. In addition, there will Program. be three interesting and informative original papers. Finally, the December issue will contain some This will be an issue for you to read and reread! reprints from early issues of the Red Cell Free Press, In addition, a letter that Dr. Tibor Greenwald including a poem by a very famous blood banker. It graciously sent to me describes some of his memories may be a good thing he concentrated on immuno- of the early days of the reference laboratory at hematology! He might be very thin now if he had kept American Red Cross National Headquarters and the to poetry! start of two rare donor programs in the United States. The first program was the Rare Donor File of the Delores Mallory American Association of Blood Banks and the second Editor-in-Chief

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 195

SPECIAL SECTION

Elution of Anti-S Hemolytic Transfusion Reaction A patient whose serum contained multiple allo A patient with hereditary hemorrhagic telangi- antibodies including anti-S was transfused with 2 units ectasia was admitted to a local hospital because of G.I. of blood that lacked all appropriate antigens except S. bleeding. The patient had a ten year history of The direct antiglobulin test post transfusion was hemoglobinuria post transfusion. Our pretransfusion weakly positive with broad spectrum and anti-IgG anti- samples were negative by standard and enzyme globulin serum but negative with anti-complement techniques. The patient received 3 units of packed reagent. The anti-S was IgG in nature, as determined by cells uneventfully. Seven days later he experienced monospecific (anti-IgG and anti-IgM) antiglobulin sera. hemoglobinuria. Serum samples tested within the next An ether eluate prepared from the patient’s post week demonstrated weak unidentified albumin- transfusion cell sample possessed no blood group antiglobulin reactivity. In an idle conversation with a antibody activity; however a heat eluate contained technologist from another Blood Center in the area, I clear cut anti-S specificity. Has anyone else experience learned that a trypsin-only anti-hr”(e) was demon- difficulty in recovering cell-bound anti-S by the ether strated in the patient’s serum 3 years previously. The elution method? We are interested in your findings. report had gone to the clinician who neglected to pass that information on to the Blood Center or transfusion M. Reid service. The patient had not been transfused since that Reference Laboratory time. Central California Regional Red Cross The patient eventually developed an identifiable Blood Program anti-Chido. The serum was tested in numerous labora- San Jose, California tories and the anti-hr”(e) could not be detected by Reprinted as published in the American Red Cross standard techniques which included many different Reference Laboratory Newletter. No. 1., October 15, enzyme techniques. The anti-hr”(e) has been detected 1976. using the microtiter technique and using the autoanalyzer. The patient has received approximately 40 units of hr”(e) negative, Chido positive blood in the last three years without any hemoglobinuria or other signs of a transfusion reaction. S. Ellisor Reference Laboratory Reprinted as published in the American Red Cross Reference Laboratory Newletter. No. 1., October 15, 1976.

Notice to Readers: All articles published, including communications and book reviews, reflect the opinions of the authors and do not necessarily reflect the official policy of the American Red Cross.

196 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

SPECIAL SECTION CONT’D

Controls for Enzyme Premodification We do not use milk flocculation or X-ray paper The following controls have proven useful in our digestion methods for quality control of enzyme laboratory for demonstrating adequate enzyme activity since they only measure proteolytic activity. premodification of red cells. M. Reid 1. Using an antibody/antigen system known to be Reference Laboratory enhanced by enzyme treatment. A weakly reactive Central California Regional Red Cross antibody, with no saline reactivity, is selected and Blood Program tested against cells possessing the weakest San Jose, California available form of the appropriate antigen. For example: Reprinted as published in the American Red Cross anti-c and heterozygous – positive cells Reference Laboratory Newletter. No. 1., October 15, 1976. anti-D and R1r cells anti-Lea and Le(a+b-) cells anti-Leb and Le(a-b+) cells 2. Using an antibody/antigen system known to be denatured by enzyme treatment. A strongly reacting antibody is selected and tested against cells possessing the strongest available form of the appropriate antigen. For example: anti-M and MM cells anti-Fya and Fy(a+b-) cells 3. Using a substance that demonstrates reduction in red cell sialic acid levels. For example: Polybrene – untreated cells aggregate Enzyme premodified cells do not aggregate Soybean extract – untreated cells do not aggregate Enzyme premodified cells aggregate

IMMUNOHEMATOLOGY IS ON THE WEB! www.redcross.org/pubs/immuno

Password “2000”

For more information or to send an e-mail message “To the editor”

[email protected]

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 197

ANNOUNCEMENTS

HEMATOLOGÍA HABANA′ 2005– Monoclonal antibodies available at no cost. The First Announcement. Laboratory of Immunochemistry at the New York The 5th National Congress and the 7th Latin American Blood Center has developed a wide range of Meeting in Hematology, Immunology, and Transfusion monoclonal antibodies (both murine and humanized) Medicine will present a scientific program at the that are useful for screening for antigen–negative International Conference Center, Havana, Cuba, May donors and for typing patients’ RBCs with a positive 16–20, 2005. A preliminary program lists malignant DAT. Monoclonal antibodies available include anti-M, hemopathies, disorders of RBC membranes, -Fya, -Fyb, -K, -k, -Kpa, -Jsb, -Dob, -Wrb, and –Rh17. For a immunotherapy, histocompatibility, immunohema- complete list of available monoclonal antibodies,please tology, hemolytic disease of the newborn, regenerative see our Web site at www.nybloodcenter.org/ medicine, and blood components as some of the framesets/FS-4C7.htm. Most of those antibodies are topics. For more information contact: Prof. José M. murine IgG and,thus,require the use of anti-mouse IgG Ballester, President, Organizing Committee, for detection, i.e, anti-K, -k, and -Kpa. Some are directly Hematology Habana′ 2005,Apartado 8070,Ciudad de la agglutinating (anti-M, -Wrb, and -Rh17), and a few have Habana, CP 10800, Cuba, e-mail: [email protected]; been humanized into the IgM isoform and are directly Web site: www.loseventos.cu/hematologia2005. agglutinating (anti-Jsb and -Fya). The monoclonal antibodies are available at no charge to anyone who requests them. Contact: Marion Reid (mreid@ Annual Symposium. The National Institutes of nybloodcenter.org) or Gregory Halverson (ghalverson@ Health, Department of Transfusion Medicine, will hold nybloodcenter.org), New York Blood Center, 310 East their 24th annual symposium, Immunohematology 67th Street, New York, NY 10021. and Blood Transfusion, on September 23, 2004. The symposium is co-hosted by the Greater Chesapeake and Potomac Region of the American Red Cross and is free of charge. Advance registration is encouraged. For more information and registration, Contact: Karen Byrne, NIH/CC/DTM, Bldg. 10/Rm. 1C711, 10 Center Drive, MSC 1184, Bethesda, MD 20892-1184; e-mail: [email protected]; or visit our Web site: www.cc.nih.gov/dtm>education.

198 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

ANNOUNCEMENTS CONT’D

Masters (MSc) in Transfusion and Transplantation Sciences At The University of Bristol, England

Applications are invited from medical or science graduates for the Master of Science (MSc) degree in Transfusion and Transplantation Sciences at the University of Bristol. The course starts in October 2004 and will last for one year. A part-time option lasting three years is also available. There may also be opportunities to continue studies for PhD or MD following MSc. The syllabus is organised jointly by The Bristol Institute for Transfusion Sciences and the University of Bristol, Department of Transplantation Sciences. It includes:

• Scientific principles underlying transfusion and transplantation • Clinical applications of these principles • Practical techniques in transfusion and transplantation • Principles of study design and biostatistics • An original research project

Applications can also be made for Diploma in Transfusion and Transplantation Science or a Certificate in Transfusion and Transplantation Science.

The course is accredited by the Institute of Biomedical Sciences.

Further information can be obtained from the Web site:

http://www.bloodnet.nbs.nhs.uk/ibgrl/MSc/MScHome.htm

For further details and application forms please contact:

Professor Ben Bradley

University of Bristol, Department of Transplantation Sciences

Southmead Hospital, Westbury-on-Trym, Bristol BS10 5NB, England

FAX +44 1179 595 342, TELEPHONE +44 1779 595 455, E-MAIL: [email protected]

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 199

ADVERTISEMENTS

NATIONAL REFERENCE LABORATORY FOR National Platelet Serology Reference BLOOD GROUP SEROLOGY Laboratory

Immunohematology Reference Diagnostic testing for: Laboratory • Neonatal alloimmune thrombocytopenia (NAIT) AABB,ARC, New York State, and CLIA licensed • Posttransfusion purpura (PTP) (215) 451-4901—24-hr. phone number • Refractoriness to platelet transfusion (215) 451-2538—Fax • Heparin-induced thrombocytopenia (HIT) American Rare Donor Program • Alloimmune idiopathic thrombocytopenia (215) 451-4900—24-hr. phone number purpura (AITP) (215) 451-2538—Fax Medical consultation available [email protected] Test methods: Immunohematology • GTI systems tests (215) 451-4902—Phone, business hours —detection of glycoprotein-specific platelet (215) 451-2538—Fax antibodies [email protected] —detection of heparin-induced antibodies (PF4 Quality Control of Cryoprecipitated-AHF ELISA) (215) 451-4903—Phone, business hours • Platelet suspension immunofluorescence test (215) 451-2538—Fax (PSIFT) • Solid phase red cell adherence (SPRCA) assay • Monoclonal antibody immobilization of platelet Granulocyte Antibody Detection and Typing antigens (MAIPA)

• Specializing in granulocyte antibody detection For information, e-mail: [email protected] and granulocyte antigen typing or call: • Patients with granulocytopenia can be classified through the following tests for proper therapy Maryann Keashen-Schnell and monitoring: (215) 451-4041 office —Granulocyte agglutination (GA) (215) 451-4205 laboratory —Granulocyte immunofluorescence (GIF) —Monoclonal Antibody Immobilization of Sandra Nance Granulocyte Antigens (MAIGA) (215) 451-4362 For information regarding services, call Gail Eiber at: (651) 291-6797, e-mail: [email protected], Scott Murphy, MD or write to: (215) 451-4877 Neutrophil Serology Reference Laboratory American Red Cross Blood Services American Red Cross St. Paul Regional Blood Services Musser Blood Center 100 South Robert Street 700 Spring Garden Street St. Paul, MN 55107 Philadelphia, PA 19123-3594

CLIA LICENSED CLIA LICENSED

200 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

ADVERTISEMENTS CONT’D

IgA/Anti-IgA Testing Reference and Consultation Services

IgA and anti-IgA testing is available to do the Red cell antibody identification and problem following: resolution • Monitor known IgA-deficient patients HLA-A, B, C, and DR typing • Investigate anaphylactic reactions • Confirm IgA-deficient donors HLA-disease association typing Our ELISA assay for IgA detects antigen to Paternity testing/DNA 0.05 mg/dL. For information regarding our services, contact For information on charges and sample Zahra Mehdizadehkashi at (503) 280-0210, or requirements, call (215) 451-4351, e-mail: [email protected], write to: or write to: Pacific Northwest Regional Blood Services American Red Cross Blood Services ATTENTION: Tissue Typing Laboratory Musser Blood Center American Red Cross 700 Spring Garden Street 3131 North Vancouver Philadelphia, PA 19123-3594 ATTN: Ann Church Portland, OR 97227

CLIA LICENSED CLIA LICENSED, ASHI ACCREDITED

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 201

Immunohematology

JOURNAL OF BLOOD GROUP SEROLOGY AND EDUCATION Instructions for Authors

SCIENTIFIC ARTICLES, REVIEWS, AND CASE REPORTS 5. Acknowledgments Before submitting a manuscript, consult current issues of Acknowledge those who have made substantial contributions to Immunohematology for style. Type the manuscript on white bond the study, including secretarial assistance; list any grants. paper (8.5" × 11") and double-space throughout. Number the pages 6. References consecutively in the upper right-hand corner, beginning with the A.In text, use superscript, arabic numbers. title page. Each component of the manuscript must start on a new B. Number references consecutively in the order they occur in page in the following order: the text. 1. Title page C.Use inclusive pages of cited references, e.g., 1431–7. 2. Abstract D.Refer to current issues of Immunohematology for style. 3. Text 7. Tables 4. Acknowledgments A.Head each with a brief title, capitalize first letter of first word 5. References (e.g., Table 1. Results of ...), and use no punctuation at the end 6. Author information of the title. 7. Tables—see 6 under Preparation B. Use short headings for each column needed and capitalize first 8. Figures—see 7 under Preparation letter of first word. Omit vertical lines. C.Place explanations in footnotes (sequence: *, †, ‡, §, ¶, **, ††). Preparation of manuscripts 8. Figures 1. Title page A.Figures can be submitted either by e-mail or as photographs A.Full title of manuscript with only first letter of first word (5″×7″ glossy). capitalized (bold title) B. Place caption for a figure on a separate page (e.g.,Fig.1.Results B. Initials and last name of each author (no degrees; all CAPS), of ...), ending with a period. If figure is submitted as a glossy, e.g., M.T.JONES and J.H. BROWN place first author’s name and figure number on back of each C.Running title of ≤ 40 characters, including spaces glossy submitted. D.3 to 10 key words C.When plotting points on a figure, use the following symbols if 2. Abstract possible: G G L L I I. A.One paragraph, no longer than 300 words 9. Author information B. Purpose, methods, findings, and conclusions of study A.List first name, middle initial, last name, highest academic 3. Key words—list under abstract degree, position held, institution and department, and 4. Text (serial pages) complete address (including zip code) for all authors. List Most manuscripts can usually,but not necessarily,be divided into country when applicable. sections (as described below). Results of surveys and review papers are examples that may need individualized sections. SCIENTIFIC ARTICLES AND CASE REPORTS SUBMITTED A.Introduction AS LETTERS TO THE EDITOR Purpose and rationale for study, including pertinent back- ground references. Preparation B. Case Report (if study calls for one) 1. Heading—To the Editor: Clinical and/or hematologic data and background serology. 2. Under heading—title with first letter capitalized. C.Materials and Methods 3. Text—write in letter format (paragraphs). Selection and number of subjects, samples, items, etc. studied 4. Author(s)—type flush right; for first author: name, degree, and description of appropriate controls, procedures, methods, institution, address (including city, state, ZIP code, and country); equipment, reagents, etc. Equipment and reagents should be for other authors: name, degree, institution, city, and state. identified in parentheses by model or lot and manufacturer’s 5. References—limited to ten. name, city, and state. Do not use patients’ names or hospital 6. One table and/or figure allowed. numbers. D.Results Presentation of concise and sequential results, referring to perti- nent tables and/or figures, if applicable. Send all submissions (original and two copies) to: E. Discussion Mary H. McGinniss, Managing Editor, Immunohematology, Implications and limitations of the study,links to other studies; 10262 Arizona Circle, Bethesda, MD 20817 and e-mail your if appropriate,link conclusions to purpose of study as stated in manuscript to Marge Manigly at [email protected] introduction.

202 IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004

Becoming a Specialist in Blood Banking (SBB)

What is a certified Specialist in Blood Banking (SBB)? • Someone with educational and work experience qualifications who successfully passes the American Society for Clinical Pathology (ASCP) board of registry (BOR) examination for the Specialist in Blood Banking. • This person will have advanced knowledge, skills, and abilities in the field of transfusion medicine and blood banking.

Individuals who have an SBB certification serve in many areas of transfusion medicine: • Serve as regulatory, technical, procedural and research advisors • Perform and direct administrative functions • Develop, validate, implement, and perform laboratory procedures • Analyze quality issues preparing and implementing corrective actions to prevent and document issues • Design and present educational programs • Provide technical and scientific training in blood transfusion medicine • Conduct research in transfusion medicine Who are SBBs? Supervisors of Transfusion Services Managers of Blood Centers LIS Coordinators Educators Supervisors of Reference Laboratories Research Scientists Consumer Safety Officers Quality Assurance Officers Technical Representatives Reference Lab Specialist Why be an SBB? Professional growth Job placement Job satisfaction Career advancement How does one become an SBB? • Attend a CAAHEP-accredited Specialist in Blood Bank Technology Program OR • Sit for the examination based on criteria established by ASCP for education and experience Fact #1: In recent years, the average SBB exam pass rate is only 38%. Fact #2: In recent years, greater than 73% of people who graduate from CAAHEP-accredited programs pass the SBB exam. Conclusion: The BEST route for obtaining an SBB certification is to attend a CAAHEP-accredited Specialist in Blood Bank Technology Program

Contact the following programs for more information: PROGRAM CONTACT NAME CONTACT INFORMATION Walter Reed Army Medical Center William Turcan 202-782-6210; [email protected] Transfusion Medicine Center at Florida Blood Services Marjorie Doty 727-568-5433 x 1514; [email protected] Univ. of Illinois at Chicago Veronica Lewis 312-996-6721; [email protected] Medical Center of Louisiana Karen Kirkley 504-903-2466; [email protected] NIH Clinical Center Department of Transfusion Medicine Karen Byrne 301-496-8335; [email protected] Johns Hopkins Hospital Christine Beritela 410-955-6580; [email protected] ARC-Central OH Region, OSU Medical Center Joanne Kosanke 614-253-2740 x 2270; [email protected] Hoxworth Blood Center/Univ. of Cincinnati Medical Center Catherine Beiting 513-558-1275; [email protected] Gulf Coast School of Blood Bank Technology Clare Wong 713-791-6201; [email protected] Univ. of Texas SW Medical Center Barbara Laird-Fryer 214-648-1785; [email protected] Univ. of Texas Medical Branch at Galveston Janet Vincent 409-772-4866; [email protected] Univ. of Texas Health Science Center at San Antonio Bonnie Fodermaier SBB Program: 210-358-2807, Linda Smith [email protected] MS Program: 210-567-8869; [email protected] Blood Center of Southeastern Wisconsin Lynne LeMense 414-937-6403; [email protected] Additional information can be found by visiting the following Web sites: www.ascp.org, www.caahep.org, and www.aabb.org

IMMUNOHEMATOLOGY, VOLUME 20, NUMBER 3, 2004 203