editor issue 11 october 2003 howard lederman, m.d. ph.d ... › sites › default › files ›...

18
Clinical Focus on Primary Immune Deficiencies ISSUE 11 OCTOBER 2003 Primary Humoral Immunodeficiency: Optimizing IgG Replacement Therapy Editor Howard Lederman, M.D. Ph.D. IDF Medical Advisory Committee Rebecca H. Buckley, M.D., Chairperson Duke University School of Medicine, Durham, NC Douglas J. Barrett, M.D. University of Florida, Gainesville, FL R. Michael Blaese, M.D. PreGentis, Inc., Newtown, PA Mary Ellen Conley, M.D. St. Jude Children’s Research Hospital, Memphis, TN Max Cooper, M.D. University of Alabama at Birmingham, Birmingham, AL Charlotte Cunningham-Rundles, M.D., Ph.D. Mount Sinai Medical Center, New York, NY Erwin W. Gelfand, M.D. National Jewish Center for Immunology and Respiratory Medicine, Denver, CO Richard Hong, M.D. University of Vermont School of Medicine, Burlington, VT Richard B. Johnston, Jr., M.D. University of Colorado School of Medicine, Denver, CO Alexander Lawton, III, M.D. Vanderbilt University School of Medicine, Nashville, TN Stephen E. Miles, M.D. All Seasons Allergy & Asthma, Woodlands, TX Hans D. Ochs, M.D. University of Washington, School of Medicine, Seattle, WA Fred Rosen, M.D. The Center for Blood Research, Boston, MA William T. Shearer, M.D., Ph.D. Texas Children’s Hospital, Houston, TX E. Richard Stiehm, M.D. UCLA School of Medicine, Los Angeles, CA John L. Sullivan, M.D. University of Massachusetts Medical Center, Worchester, MA Diane W. Wara, M.D. UCSF Medical Center, San Francisco, CA Jerry Winkelstein, M.D. Johns Hopkins University School of Medicine, Baltimore, MD Jonathan C. Goldsmith, M.D. IDF Vice President of Medical Affairs, Towson, MD

Upload: others

Post on 29-Jun-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Editor ISSUE 11 OCTOBER 2003 Howard Lederman, M.D. Ph.D ... › sites › default › files › ... · have defective humoral immunity with normal cell-mediated immune function,or

Clinical Focuson Primary Immune Deficiencies

ISSUE 11 OCTOBER 2003

Primary HumoralImmunodeficiency: Optimizing IgG Replacement Therapy

EditorHoward Lederman, M.D. Ph.D.

IDF Medical Advisory CommitteeRebecca H. Buckley, M.D., ChairpersonDuke University School of Medicine, Durham, NC

Douglas J. Barrett, M.D.University of Florida, Gainesville, FL

R. Michael Blaese, M.D.PreGentis, Inc., Newtown, PA

Mary Ellen Conley, M.D.St. Jude Children’s Research Hospital,Memphis, TN

Max Cooper, M.D.University of Alabama at Birmingham,Birmingham, AL

Charlotte Cunningham-Rundles, M.D., Ph.D.Mount Sinai Medical Center, New York, NY

Erwin W. Gelfand, M.D.National Jewish Center for Immunology andRespiratory Medicine, Denver, CO

Richard Hong, M.D.University of Vermont School of Medicine,Burlington, VT

Richard B. Johnston, Jr., M.D.University of Colorado School of Medicine,Denver, CO

Alexander Lawton, III, M.D.Vanderbilt University School of Medicine,Nashville, TN

Stephen E. Miles, M.D.All Seasons Allergy & Asthma, Woodlands, TX

Hans D. Ochs, M.D.University of Washington, School of Medicine,Seattle, WA

Fred Rosen, M.D.The Center for Blood Research, Boston, MA

William T. Shearer, M.D., Ph.D.Texas Children’s Hospital, Houston, TX

E. Richard Stiehm, M.D.UCLA School of Medicine, Los Angeles, CA

John L. Sullivan, M.D.University of Massachusetts Medical Center,Worchester, MA

Diane W. Wara, M.D.UCSF Medical Center, San Francisco, CA

Jerry Winkelstein, M.D.Johns Hopkins University School of Medicine,Baltimore, MD

Jonathan C. Goldsmith, M.D.IDF Vice President of Medical Affairs,Towson, MD

Page 2: Editor ISSUE 11 OCTOBER 2003 Howard Lederman, M.D. Ph.D ... › sites › default › files › ... · have defective humoral immunity with normal cell-mediated immune function,or

IGIVs (Immune Globulin

Intravenous (Human)) are all

manufactured differently –

but how does that impact a

product’s efficacy or tolerability?

The medical community now

expects solid evidence-based

medicine through:

• controlled and comparative

trial designs

• well-defined clinical

endpoints

• appropriate trial size

At Bayer, we agree.

Bayer is taking a

New Perspective.

Are a l l IGIVs

the same?

Cre

ativ

e by

Eur

o RS

CG

Life

Tor

onto

what ’s in IGIV?

nex

t

www.gamunex.comwww.IGIVnext .com

Page 3: Editor ISSUE 11 OCTOBER 2003 Howard Lederman, M.D. Ph.D ... › sites › default › files › ... · have defective humoral immunity with normal cell-mediated immune function,or

Erwin W. Gelfand1, M.D.,

Jonathan Goldsmith2, M.D., and

Howard M. Lederman3, M.D., Ph.D.

1Division of Cell Biology, Department of

Pediatrics, National Jewish Medical and Research

Center, 1400 Jackson Street, Denver, CO 80206;2Immune Deficiency Foundation,

40 W. Chesapeake Avenue, Suite 308,

Towson, MD 21204; and 3Eudowood Division of Pediatric Allergy and

Immunology, Johns Hopkins Hospital, 600 N.

Wolfe Street, CMSC 1102, Baltimore, MD 21287

Primary immune deficiency diseases (PIDD) are

naturally occurring defects of the immune system

and comprise a diverse group of illnesses. In the last

decade, incredible progress has been made in the

diagnosis and treatment of these diseases. Concerns

remain about under diagnosis but important

educational advances have certainly contributed to

a dramatic increase in awareness and suspicion of

PIDD in the general medical community. Continuing

efforts to ensure that patients are diagnosed and

treated as early as possible remain a priority.

Importantly, rapid advances in defining the genetic

bases of the diseases, the potential for antenatal

diagnosis, and even prospects for successful gene

therapy have all been accomplished.To date, 150-200

genetic defects have been defined (1).

Among the patients with PIDD are a group of

disorders in which the ability to produce antibody

is reduced or absent (Table 1). Such problems can

be caused by defects intrinsic to the B lymphocyte

(e.g., X-linked agammaglobulinemia in which

B lymphocytes fail to mature) and by problems

intrinsic to the CD4 helper T lymphocyte (e.g.

X-linked Hyper-IgM syndrome). Patients may

have defective humoral immunity with normal

cell-mediated immune function, or defects of both

arms of the immune system.

Increased susceptibility to infection is common to

all of the primary humoral immunodeficiencies,

with the possible exception of asymptomatic IgA

deficiency.With the rapid institution of antibiotics,

it is rare today to see osteomyelitis, meningitis or

consolidated pneumonia as the presenting feature.

More commonly, recurrent upper respiratory tract

infections, such as otitis media and sinusitis are

seen. Less commonly, patients develop mastoiditis,

failure to thrive and chronic/recurrent diarrhea. In

some forms of antibody deficiency, such as the

hyper-IgM syndrome, autoimmune cytopenias, or

infection with unusual pathogens such as

Pneumocystis carinii or Cryptosporidium parvum

infection, and chronic inflammatory disorders

(e.g., sclerosing cholangitis) may be features of the

disease.

October 2003 Immune Deficiency Foundation: Clinical Focus 3

Defects that exclusively impair humoral immunity• X-linked agammaglobulinemia• Selective IgA deficiency• IgG subclass deficiency

Defects that predominantly impair humoral immunity• Common variable immunodeficiency• X-linked hyper-IgM syndrome• Wiskott-Aldrich syndrome

Defects of both humoral and cell-mediated immunity• Severe combined immunodeficiency• DiGeorge Syndrome

Primary Humoral Immunodeficiency:Optimizing IgG Replacement Therapy

Table 1: Examples of PrimaryImmunodeficiency DiseasesThat Impair Humoral Immunity

Page 4: Editor ISSUE 11 OCTOBER 2003 Howard Lederman, M.D. Ph.D ... › sites › default › files › ... · have defective humoral immunity with normal cell-mediated immune function,or

The most common offending organisms generally

are encapsulated bacteria, such as Hemophilus

influenzae and Streptococcus pneumoniae.

Patients with humoral immunodeficiency also

appear to be uniquely susceptible to systemic

mycoplasma infection, especially Ureaplasma

urealyticum, M. hominis and other species (2-4).

Indeed, some of the arthritis and osteomyelitis

seen in these patients may be secondary to

mycoplasma infection (2, 5). Similarly, acute and

chronic lung disease may also be due to these

organisms (3, 4). Often, the mycoplasma infections

are insidious, evoking only a low grade febrile and

leukocyte response.These organisms are very

difficult to culture and do not respond to the

usual antibiotics instituted for infections caused by

encapsulated organisms.Tetracyclines or quinolones

are often required to eradicate these organisms (2,5).

The foundation for therapy of the humoral immun-

odeficiency diseases is immunoglobulin (IgG)

replacement.This form of therapy has evolved

significantly over the last 50 years. Bruton, in 1952,

first treated his patient with subcutaneous injec-

tions of gammaglobulin. Intramuscular injections

prevailed for almost 30 years until Bayer

Pharmaceuticals introduced the first ready-to-use

liquid preparation of modified gammaglobulin

suitable for intravenous infusion in North America

in the early 1980’s. Intramuscular gammaglobulin

was generally injected at 0.6 cc (100 mg)/kg body

weight.These injections were painful and limited

the amounts injected. Nonetheless, even at these

doses, reports of benefit emerged with reduction

in febrile episodes and the incidence of infection.

With the introduction of intravenous gammaglobulin

replacement (IGIV), therapeutic strategies also

changed.The ability to administer IgG intravenously

provided the possibility to administer larger

amounts. Several studies demonstrated the benefit

of higher doses (400-600 mg/kg body weight) in

infection prophylaxis and improvement in lung

function (6, 7). Furthermore, the higher doses were

associated with elimination of mycoplasma (8). It

appeared that maintenance of trough levels

(4 weeks post-infusion) above 500 mg/dL resulted

in a reduction of major and minor infections, and

the need for hospitalization.These studies also

revealed individual variations in time to achieve a

stable plateau at trough and in catabolic rates.

These findings highlight the need to tailor IGIV

regimens to the requirements of the individual

patient and that a single fixed dose regimen may

not be applicable to all patients.

Despite almost five decades of use and more than

two decades of IGIV therapy, there is little docu-

mentation, which specifically addresses the efficacy

of IGIV in the prevention of significant infections,

especially pneumonia, and long-term and progressive

lung disease.There has been reasonable documen-

tation of the overall effectiveness of IGIV in

preventing acute respiratory infections, otitis, and

sinusitis, but some patients still suffer from these

and gastrointestinal problems and other infections,

despite institution of IGIV prophylaxis. Replacement

IgG therapy, at standard doses, may not interrupt

the occurrence or progression of permanent lung

damage (9). Acute pulmonary infections may be

significantly prevented, but not permanent lung

damage (10). Patients who present with significant

lung disease, such as bronchiectasis,prior to initiation

of IGIV therapy, may continue to suffer from

progressive lung disease. In turn, a subset of patients,

despite optimal IGIV dosing and maintenance of

adequate trough levels, may also develop permanent

lung damage for reasons that are not currently

understood. Chronic lung disease in patients with

primary humoral immunodeficiency remains a

major factor affecting quality of life and longevity.

At present, it is not known why “therapeutic failures”

continue to occur despite the many advances in

treatment. Surprisingly, in a recent survey carried

out by the Immune Deficiency Foundation (IDF),

40% of patients felt that their disease was not optimally

controlled and recurrent infections persisted.

Approximately 60% complained that the effects of

4 Immune Deficiency Foundation: Clinical Focus October 2003

Page 5: Editor ISSUE 11 OCTOBER 2003 Howard Lederman, M.D. Ph.D ... › sites › default › files › ... · have defective humoral immunity with normal cell-mediated immune function,or

IGIV were wearing off prior to their next scheduled

infusion, even two weeks after an infusion.There

are several possibilities to explain these incomplete

responses, suggesting the need to regularly re-evaluate

our approach to these patients.

Management GuidelinesAlthough the data may not be at hand, it makes

empirical sense that the earlier the diagnosis and

institution of appropriate therapy, the better the

likelihood of preventing permanent lung damage.

It is unclear if there are significant numbers of

primary humoral immunodeficiency patients who

are undiagnosed, who warrant therapy or whose

therapy has been delayed. It is not surprising that

the diagnosis may be delayed given the usual find-

ings today that these patients most often present

with common infections, such as otitis media and

sinusitis, and initially respond appropriately to

routine antibiotics. Indeed, a common presenting

symptom in adults may be fatigue with little in the

way of significant infections.

Once the diagnosis of (antibody) immunodeficiency

is made, it is important to establish, at baseline, the

extent of lung damage.The most effective way is to

perform a high-resolution computed tomography

(CT) scan of the chest (9). If lung damage, e.g.,

bronchiectasis, is detected at presentation, then

management may need to be modified. Follow-up

CT lung scans should be considered every 12-24

months, depending on the presenting features.

Similarly, an initial CT scan of the paranasal sinuses

may aid in the long-term management of these

patients (Table 2).

The recent IDF survey also identified that 71% of

patients were followed by primary care physicians

and not by an immunologist. Primary humoral

immunodeficiency is a lifelong and chronic disease

requiring close co-operation between the patient,

the primary care physician and the specialist.

AntibioticsA mainstay of therapy of antibody deficiency disorders

is appropriate use of antibiotics.As discussed,

patients with antibody deficiency disorders are

susceptible to the common encapsulated bacteria

(H. influenzae, S. pneumoniae), but also are uniquely

susceptible to mycoplasma.The former are easy to

culture, the latter are not and the specific antibiotics

for each are quite different. Because of the difficulty

in isolating the mycoplasmas, empirical therapy

(e.g., doxycycline) may be indicated in patients

with chronic/recurrent infection, who fail to

respond to usual antibiotics.

IgG ReplacementIGIV is the current standard of therapy with few if

any indications currently for intramuscular injections.

Most patients receive IgG replacement via the

intravenous route although some patients receive

subcutaneous infusions (11). Suboptimal clinical

outcomes could reflect a number of possibilities.

Although there are no obvious differences in efficacy,

careful scrutiny must be exercised as route and

frequency of administration could potentially affect

outcome. For example, it is not clear whether more

frequent infusions at lower doses are more or less

effective than larger infusions every three to four

weeks.After appropriate training and the absence

October 2003 Immune Deficiency Foundation: Clinical Focus 5

Table 2: Guidelines for Managing Patients with AntibodyDeficiency

• An immunologist should direct life-long care of the patient

• Assess pulmonary status with high resolution computed tomography (CT) and spirometry at baseline and every 12-24 months

• IgG replacement therapy at doses of at least 400-mg/kg/month and higher doses in patients whereinfections persist, especially bronchiectasis

• Institute appropriate antibiotic therapy when indicated

Page 6: Editor ISSUE 11 OCTOBER 2003 Howard Lederman, M.D. Ph.D ... › sites › default › files › ... · have defective humoral immunity with normal cell-mediated immune function,or

of a history of adverse reactions, infusion by either

route can be performed at home. If monitored

carefully, home infusion programs can be as effective

as monthly visits to a recognized center under

physician supervision. Convenience, however, is not

a substitute for good medical management and home

infusion programs do not replace the requirement

for regular patient follow-up with physicians.

From the IDF survey, it appears that a significant

proportion of patients may be under-dosed. Many

of the product package inserts recommend monthly

doses of 100-200 mg/kg body weight, but numer-

ous studies have convincingly shown how dose

and trough levels affect outcome. In a double

blind, randomized study in PIDD patients, doubling

the dose of IGIV (from 300 to 600 mg/kg body

weight in adults and 400 to 800 mg/kg body weight

in children) significantly reduced the number and

duration of infections (12).Appropriate dosing is

essential and should not be determined by costs,

longer infusion times,or concerns about convenience.

Mechanism of Action of IgG ReplacementIt is interesting to speculate on how IgG replacement

works in the antibody deficiency diseases. Is it

simply replacement of missing antibodies or do

other potentially important activities come into

play? For many organisms, the levels of specific

antibodies are surprisingly low (e.g., against common

serotypes of S.pneumoniae) and are not standardized

from batch-to-batch or brand-to-brand (see below).

Interestingly, there are virtually no detectable anti-

bodies to the mycoplasmas, yet IGIV appears to

play a role in their elimination or containment.

IGIV has also been shown to have a number of

anti-inflammatory and immunomodulatory activities

(13).These include inhibition of the production of

pro-inflammatory cytokines, neutralization of

bacterial exotoxins, modulation of Fcg receptors,

triggering of lymphocyte apoptosis and prevention

of deposition of the complement membrane attack

complex.Whether any of these or other activities

of IGIV play a role in these patients beyond

antibody replacement is an interesting possibility.

Are All IGIVs the Same?When surveyed,patients express four major concerns

about IGIV: safety, supply, tolerability/adverse events,

and outcome. For the patients, their physicians and

other healthcare personnel, there has been an

assumption that all eight licensed products in the

United States are equivalent in these and other

parameters.This assumption has in large part been

fostered by the absence of any significant comparison

data. However, these products do differ in terms of

donor pools, manufacturing and final formulation.

It is possible that a number of these differences

can affect tolerability, risk of adverse events,

infusion rate and efficacy.

Differences in basic fractionation and the addition

of various modifications for further purification,

stabilization and virus inactivation/ removal have

yielded products clearly different from one to the

other (Table 3).There are well-established differences

in chemical structure, antibody content, subclass

distribution and electrophoretic profile. Further,

the composition of the final product also differs

widely. Some attempts at standardization, for

example by a WHO Expert Committee on Biologic

Standardization (14), has mandated that the IgG

be as unmodified as possible, maintain its biologic

function (opsonic activity, complement fixation,

Fc-receptor binding), contain certain levels of specific

antibody and meet accepted safety standards.

Despite meeting these standards, it is possible that

the different preparations and the modifications

used to enable safe intravenous administration can

induce alterations in the biologic activity of the

IgG molecule (15, 16).Whereas most commercial

preparations are screened for levels of antibodies

to several viral and bacterial antigens to validate

purification procedures, there is little routine

evaluation of the relationship between antibody

6 Immune Deficiency Foundation: Clinical Focus October 2003

Page 7: Editor ISSUE 11 OCTOBER 2003 Howard Lederman, M.D. Ph.D ... › sites › default › files › ... · have defective humoral immunity with normal cell-mediated immune function,or

October 2003 Immune Deficiency Foundation: Clinical Focus 7

Polygam S/D5% 10%

Panglobulin Gammar-P I.V. Gammagard S/D5% 10%

Iveegam EN Gamunex1 Venoglobulin-S5% 10%

Carimune NF

American Red Cross

American Red Cross

Aventis Behring, LLC

Baxter Corporation/BioScience Division

Baxter Corporation/BioScience Division

Bayer HealthCare/

Biologic Products Division

Grifols ZLB Bioplasma, Inc.

Cohn-Oncley fractionation,

ultra-filtration,ion-exchange

chromatography,solvent

detergent treatment

Kistler Nitschmann

fractionation,pH 4.0,

trace pepsin,nanofiltration

Cohn-Oncley fractionation,ultra-filtrationpasteurization

at 60° C for 10 hours

Cohn-Oncley fractionation,

ultra-filtration,ion-exchange

chromatography,solvent

detergent treatment

Cold ethanol fractionation,PEG, trypsin treatment

Cohn-Oncley fractionation,

caprylate/ chromatography

purification,cloth and depth

filtration,final container

low pH incubation

Cold alcohol fractionation,PEG/Bentonite fractionation,ion-exchange

chromatography,solvent

detergent treatment

Kistler Nitschmann

fractionation,pH 4.0,

trace pepsin,nanofiltration

Lyophilized Lyophilized Lyophilized Lyophilized Lyophilized Liquid Liquid Lyophilized

24Months

24 Months

24 Months

24 Months

24 Months

36 Months

24Months

24 Months

<5 minutes at roomtemperature >20 minutes

if cold

Several minutes

<20 minutes <5 minutes at roomtemperature >20 minutes

if cold

≤10 minutes at room

temperature

None (Liquid Solution)

None(Liquid Solution)

Several minutes

5% 10% 3 to 12% 5% 5% 10% 5% 10% 5% 10% 3 to 12%

4 mL/ 8 mL/kg/hour kg/hour

>2.5 mL/kg/hour 3.6 mL/kg/hour 4 mL/ 8 mL/ kg/hour kg/hour

1.8 mL/kg/hour 4.8 mL/kg/hour 4.8 mL/ 3.0 mL/kg/hour kg/hour

>2.5 mL/kg/hour

2.5 0.6 hours hours

<3.3 hours(6% solution)

2.8 hours 2.5 0.6hours hours

5.6 hours 1.0 hours 2.1 1.7 hours hours

<3.3 hours (6% Solution)

20 mg/mL 40 mg/mLglucose glucose

1.67 gm sucrose pergram of protein

50 mg/mL sucrose 20 mg/mL 40 mg/mLglucose glucose

50 mg/mL glucose None 50 mg/mL D-Sorbitol

1.67 gm sucrose pergram of protein

8.5 mg/mL 17 mg/mLsodium sodiumchloride chloride

<20 mg sodiumchloride per

gram of protein

5 mg/mLsodium chloride

8.5 mg/mL 17 mg/mLsodium sodiumchloride chloride

3 mg/mLsodium chloride

TraceAmounts

<1 mEq/L

<20 mg sodiumchloride per

gram of protein

636 1250mOsm/L mOsm/L

192 - 1074mOsm/kg

309 mOsm/L

636 1250mOsm/L mOsm/L

≥240 mOsm/L 258 mOsm/kg 300 330mOsm/L mOsm/L

192 - 1074mOsm/kg

6.4 - 7.2 6.4 - 6.8 6.4 - 7.2 6.4 - 7.2 6.4 - 7.2 4.0 - 4.5 5.2 - 5.8 6.4 - 6.8

< 2.2 µg/mL in a 5% solution

720 µg/mL <25 µg/mL < 2.2 µg/mL in a 5% solution

<10 µg/mL 46 µg/mL 15.1 20 - 50 µg/mL µg/mL

720 µg/mL

Table 3: Characteristics of IGIV Products Licensed for Use in the United States (October 2003)

BRANDNAME

Manufacturer or Distributor

Method ofProduction(Including

Viral Inactivation)

Form

Shelf-Life

ReconstitutionTime

Maximum RecommendedInfusion Rate

Time to Infuse 35 gms2

Sugar Content

Sodium Content

Osmolarity/Osmolality

PH

IgA Content

AvailableConcentrations

1 Gamunex will replace Gamimune N, 10% during the first half of 2004.2 0.5 gm/kg for a 70 kg adult = 35 gms; 5% Concentrations: 1g = 20 mL; 10% Concentrations: 1g = 10 mL

The time to infuse is based on the maximal infusion rate.

Page 8: Editor ISSUE 11 OCTOBER 2003 Howard Lederman, M.D. Ph.D ... › sites › default › files › ... · have defective humoral immunity with normal cell-mediated immune function,or

titer and antibody function. Evaluation by determi-

nations of antibody avidity or affinity, opsonic

activity or viral neutralization that may be affected

by the purification steps, is rarely carried out.

Comparative studies have shown differences and

lack of consistency (product-to-product/batch-to-

batch) that affect (reduce) opsonic activity (17-19).

These and other differences related to enzyme

treatment, chemical modification and fractionation,

underscore the need for concerns about biologic

efficacy and not simply meeting the standards

established for protein electrophoretic profile and

subclass determinations.These differences have also

been extended to the protective effects of IGIV in

patients with antibody deficiency (20).Although

the commercial preparations are different with

varying biologic activities, no systematic evaluation

of the clinical implication of this fact has been

reported, making it difficult for the clinician to

select the best preparation for a specific indication.

Is there an “ideal” IGIV? The concept of ideal may

vary significantly depending on the constituency

queried – manufacturers require product integrity,

the clinician is primarily interested in efficacy,

while the patient may identify safety as the critical

feature. For pharmacists, often in a central decision-

making position, the acquisition cost of a product

may be the major determining factor in product

selection. However, selection based on acquisition

cost alone may be more costly in the long run if

biologic function or efficacy is compromised or

adverse events are higher and need to be managed.

Given the paucity of comparative trials or data, what

factors or criteria may be used to help the clinician

choose among the many products available? The

factors include manufacturing process, safety,

composition of the final product, tolerability and

efficacy.As a consequence, many of the defined

functions may also differ, as may a large number

of the unknowns that contribute to efficacy in

the wide-ranging number of diseases now being

treated with IGIV.

Production of IGIVIGIVs are prepared from plasma pooled from

thousands of donors. Most production processes

begin with sequential precipitation and fractionation

with ethanol to isolate IgG from other plasma proteins.

The material is subjected to freeze-drying to remove

the ethanol and produce stable intermediates.

Freeze drying in the presence of ethanol promotes

formation of insoluble IgG aggregates.The IgG

concentrates from initial fractionation are then

subjected to additional processing to produce

material suitable for intravenous administration.

This is where major differences exist among

products and where biologic function is most

susceptible to alteration.Treatment with proteolytic

enzymes generally gave way to chemical modification

in an attempt to preserve the integrity of the IgG

molecule, reduce aggregate formation and eliminate

anti-complementary activity. Further modifications

led to improved products, higher purity, improved

stability and normal IgG subclass distribution.

Because of the different modifications used, it is

not surprising that the different products vary

when the amounts of IgG monomer, dimer and

polymers are assessed, fragment levels are examined

and the levels of excipient proteins are quantified

(e.g., albumin).While they can vary from lot to lot

within a product, the manufacturer’s specifications

must be met. It is not unreasonable to assume that

longer process times, more suspension/precipitation

steps, and harshness of the treatment procedures

contribute to some loss of the integrity of the IgG

molecules and biologic function as well as create

the potential for differences in efficacy, incidence

of adverse events and batch-to-batch variability.

A major goal in improving the available products

would be to increase yield (to help overcome

supply issues), reduce processing time, minimize

the harshness of the modification procedures, and

ensure purity while maintaining batch-to-batch

consistency.

8 Immune Deficiency Foundation: Clinical Focus October 2003

Page 9: Editor ISSUE 11 OCTOBER 2003 Howard Lederman, M.D. Ph.D ... › sites › default › files › ... · have defective humoral immunity with normal cell-mediated immune function,or

Product CharacteristicsLiquid vs. LyophilizedThe manufacturing process also impacts other

important features.The first is whether the final

product is in liquid form or not. Liquid preparations

have been accepted as more convenient, easier to

use and may be associated with fewer adverse

events. In ready-to-use form, the liquid preparations

shorten preparation time and delays for patients.

When there is concern for wastage for example, if

the patient does not show up for an appointment,

liquid products can be returned to inventory and

used later. Concerns about wastage often mean

that preparation of lyophilized products does not

begin until the patient arrives.

Product ConcentrationThe second consequence of the manufacturing

process is product concentration. Products that

can be given at higher concentrations decrease

volume load, an important aspect in certain patient

populations. For example, a 40 kg patient receiving

0.5 gm IGIV/kg body weight would receive 200 mL

of a 10% solution compared to 400 mL of a 5%

solution. Simply concentrating certain products by

reconstitution in a smaller volume will increase the

osmolality of the final solution and may contribute

to significant adverse events such as renal compli-

cations or thromboembolic episodes (see below).

Viral InactivationMinimizing the risk of transmission of an infectious

disease is now required during the manufacturing

of IGIV. Safety standards have been implemented

with respect to viral pathogens, including

documentation of the capacity of the manufacturing

process to remove or inactivate viruses. Plasma

testing, both of individual donations using serologi-

cal tests and manufacturing pools using poly-

merase chain reaction tests, is the initial step in

providing source material that is free of high levels

of clinically significant viruses.Accepted viral

inactivation or removal steps include treatment

with solvent detergent, polyethylene glycol (PEG),

enzyme treatment including pepsin or trypsin,

pasteurization, caprylate, acidification, PEG

Bentonite, nanofiltration and depth filtration.

Traditional calculation of overall process reduction

of viruses has been based on the sum of reductions

determined for individual production steps (21).

An important aspect is the selection of comple-

mentary safety steps for incorporation into the

manufacturing process. If these steps act through

independent mechanisms in the inactivation/removal

of viruses, they can be considered additive, thereby

increasing overall safety and providing the widest

possible safety margin against known and unknown

viruses. Further validation of the robustness of these

viral reduction steps, under conditions outside

standard operating conditions adds to the safety

margin.

Recently, a new IGIV product has been licensed:

Gamunex, Immune Globulin Intravenous (Human),

10% Caprylate/Chromatography Purified.The

production process for Gamunex includes viral

inactivation with the use of caprylate (caprylic

acid), a naturally occurring octanoic fatty acid.

Caprylate has been shown to be an effective virus

inactivator with rapid kinetics and robustness

when evaluated at different pH, temperature and

protein concentrations (22).The kinetics of virus

inactivation are much faster than seen with solvent

detergent under many conditions.As time in the

manufacturing process is a critical variable for quality

of the end product, introduction of caprylate may

not only be less harsh to the IgG molecule than

solvent detergent or pasteurization, but could

markedly shorten processing time.This shorter

processing time could impact the quality of the

end product.

Advances have also been made in effective

screening for prion contamination on an

experimental basis. Once fully evaluated, these

methods may permit direct testing of the

inactivation steps for reduction or elimination of

the potential for prion contamination.

October 2003 Immune Deficiency Foundation: Clinical Focus 9

Page 10: Editor ISSUE 11 OCTOBER 2003 Howard Lederman, M.D. Ph.D ... › sites › default › files › ... · have defective humoral immunity with normal cell-mediated immune function,or

Product CompositionAs discussed in a NIH consensus conference on

IGIV (23), the variety of manufacturing processes,

as well as starting materials, leads to differences

among preparations that may be clinically important.

Choosing the preparation of IGIV must take into

account specific differences that can significantly

impact the outcome in recipients (Table 4).

Fluid VolumesIssues related to fluid load were discussed above

and are linked to the final concentration of the

solution.The ability to deliver higher amounts of

IgG in lower volumes has a major impact on

recipients who may be intolerant of large fluid

volumes, such as infants or patients with congestive

heart failure or renal insufficiency. In addition, larger

fluid volumes require longer infusion times, a

factor that patients may like to minimize.

Sugar ContentVarious sugars (Table 3), sorbitol, glucose and

sucrose have been added to some preparations as

a stabilizer, preventing aggregate formation. Some

products contain no sugar.The major problem

associated with sugar content is the incidence of

significant adverse events, particularly acute renal

failure or insufficiency.Although rare, the CDC

reported that 90% of the IGIV-associated renal

adverse events in the United States occurred with

sucrose-containing IGIV preparations (24).

Sodium ContentIn the commercially available preparations, sodium

content varies widely from trace amounts to 0.85%

concentrations (Table 3). Caution must be exercised

when lyophilized preparations are reconstituted

to higher concentrations in an attempt to reduce

volume load. In some instances, concentrating a

lyophilized product from 5% to 10% can create a

nearly 2% saline solution. Concerns about increased

salt concentrations and association with significant

adverse events and thromboembolic complications

have been raised.

OsmolalityIn IGIV solutions, the major contributors to

osmolality include sodium, sugars, and other

excipient proteins. Physiologic osmolality is 280-

296 mOsm/kg of water. Solutions of IGIV range

from physiologic osmolality to solutions that far

exceed these levels, greater than 1,000 mOsm.

Some sugar-stabilized products have higher

osmolalities than sugar-free preparations. In

reconstituting lyophilized preparations, careful

attention to osmolality is required as significant

adverse events may occur with solutions exceeding

the physiologic range.With some lyophilized

preparations, reconstitution to higher concentrated

solutions results in hyperosmolar solutions.

pHThe pH optimum for IGIV to prevent aggregation

is 4.0-4.5.As a consequence, for preparations at

higher pH, agents are added to maintain stability

and prevent aggregation.There are scattered reports

that low pH may be associated with phlebitis.

IgA ContentPatients with selective IgA deficiency and the

ability to produce antibodies may be at risk for

10 Immune Deficiency Foundation: Clinical Focus October 2003

• Final preparation in liquid or lyophilized form

• IgG concentration

• Viral inactivation process

• Sugar content

• Sodium content

• Osmolality

• pH

• IgA content

Table 4: Differences Among IGIVPreparations

Page 11: Editor ISSUE 11 OCTOBER 2003 Howard Lederman, M.D. Ph.D ... › sites › default › files › ... · have defective humoral immunity with normal cell-mediated immune function,or

developing IgE or IgG anti-IgA antibodies resulting

in reactions.Although anaphylaxis is a theoretical

risk, it is indeed very rare; so rare that the NIH

consensus conference did not recommend

screening for anti-IgA antibodies in IGIV recipients

(23).The content of IgA in a given preparation,

except in the rarest of circumstances, is not usually

an important factor.

Isohemagglutinin TitersPreparations of IGIV do contain low-titered anti-A,

anti-B, and anti-Rh blood group antibodies and

they may be detectable, transiently, in post-treatment

direct and indirect antiglobulin tests (25).There

are no known reports of these antibodies in

(non-hyperimmune) IGIV preparations being

associated with hemolysis.The European Union

Pharmacopea mandates that anti-A and anti-B titers

be less than 1:64 in IGIV preparations.

Antibody TitersThere are marked differences in the levels of some

antibodies among different preparations. Levels of

certain antibodies, e.g., to tetanus or to ubiquitous

organisms, such as H. influenzae type b, may not

differ significantly. However, realization of such

differences has prompted screening of certain

preparations for higher titers to treat specific

diseases, as for example, chronic ECHO virus

meningitis in antibody-deficient patients (26).

Differences in antibody titers can influence clinical

outcomes as was shown in clinical trials of low

birth weight neonates treated with IGIV – here

antibody levels against S. epidermidis were an

important factor in determining success or failure

(27).As antibody levels may also play a role in the

mechanism of action of IGIV in PIDD or different

autoimmune and allergic diseases (e.g., anti-idio-

type, anti-exotoxin/superantigen, anti-cytokine

levels, etc.), these differences in antibody content

between preparations could significantly determine

efficacy of intervention with IGIV.

TolerabilityTolerability, the ability to receive IGIV without

incident and at rates that reduce the need for

burdensome and long infusion times, varies

markedly among preparations. In patients with

primary immune deficiency diseases, the incidence

of adverse events ranges from <5-16%, whereas

the incidence of adverse events is higher among

patients with immune-mediated diseases, such as

immune thrombocytopenia.This may be related to

differences in IGIV dose that is generally 0.5 gm/kg

for immunodeficiency, but 1-2 gm/kg for immune-

mediated diseases.The reactions most commonly

seen include headache, fever, myalgia, chills, nausea

and vomiting.The cause(s) of these reactions is not

known but may be the result of aggregate formation.

Many patients and clinicians notice differences

among products in tolerability related to headache,

fever, chills and shortness of breath.Transient

elevations of serum ALT and AST without clinical

correlation have been seen in some patients

following infusions of different preparations of

IGIV (28,29). Issues related to tolerability were

significant enough to trigger a switch to another

product in 24% of patients, while another 18%

either refused a product or delayed the infusion

in the recent IDF survey.

An important issue in the administration of IGIV is

the rate of infusion as this relates directly to patient

acceptability.The incidence of adverse events,

including thromboembolic events, has been tied to

the rate of infusion (30, 31). Current recommended

rates fall within the range of 0.03-0.13 mL/kg/min,

depending on the preparation.There are in fact

relatively few studies examining the tolerability of

rapid infusion of IGIV.The concentration of the

solution and the rate will dictate the length of time

an infusion will take (Table 3).With a 10% solution

at a rate of 0.08 mL/kg/min (8 mg/kg/min, 480

mg/kg/hr), an infusion of 0.5 gm/kg would require

about one hour to complete compared to a 5%

solution, which would require two hours. Some

IGIV preparations have slower recommended rates

of infusion.

October 2003 Immune Deficiency Foundation: Clinical Focus 11

Page 12: Editor ISSUE 11 OCTOBER 2003 Howard Lederman, M.D. Ph.D ... › sites › default › files › ... · have defective humoral immunity with normal cell-mediated immune function,or

EfficacyUltimately, efficacy is a key and primary concern

dictating selection of a particular product.There

have been few, if any, comprehensive comparisons

of efficacy made in a controlled, clinical trial setting.

Recently, a direct comparison between products

was made. In PIDD patients, infusion of Gamimune N

10% at equivalent doses and frequency of infusions

was compared to infusion of a new product,

Gamunex 10%.This multi-center clinical trial was

unprecedented in many ways.More than 170 patients

were enrolled compared to many previous licensure-

relevant trials that enrolled 15-50 patients.This trial

had clearly defined and clinically relevant end-points.

The new product was manufactured by an entirely

new process, which deliberately avoided harsh

detergents; shortened production time (by 70%);

and increased the efficiency of the entire process.

Caprylate was used for rapid virus inactivation and

as an additional purification step.The trial was

powered to demonstrate non-inferiority showing

that Gamunex was at least as effective as

Gamimune N 10%.Yet, Gamunex showed in virtually

all end-points, including incidence of validated

infections, surprising differences in efficacy that

challenge current perceptions that all products are

expected to provide similar clinical outcomes. In

particular, the incidence of validated infections was

reduced to a level not previously reported in antibody-

deficient PIDD patients (32). Controlled trials such

as this one suggest that some differences in

preparation and formulation might affect clinical

outcomes. Confirming studies are needed to

learn more about this intriguing possibility.

How these differences potentially affect functional

activity of the IgG molecule, IgG circulating half-life

or other biologic functions remains to be determined.

Conclusions and Recommendations1. Early diagnosis and institution of appropriate

antibiotic and IGIV therapy are necessary to

minimize long-term sequelae in PIDD patients

with antibody deficiency.

2.Education of primary care physicians,pediatricians,

internists, and otolaryngologists is necessary to

minimize delays in diagnosis.

3. An integrated team approach, including the primary

physician, specialist, infusionist, pharmacist, and

the patient is necessary to optimize care.

4. Management can be optimized by establishing

the level of baseline lung and sinus disease with

CT scans and lung function tests and establishing a

plan for regular follow-up to prevent lung disease

progression.

5. Prompt institution of appropriate antibiotic

therapy, especially in patients with chronic or

recurrent disease is an important part of treatment.

6.The importance of appropriate dosing regimens

specific to the needs of the individual patient

should be recognized.

7. Differences among IGIV products (tolerability,

risk of adverse effects, recommended rates of

infusion and potentially efficacy) must be coupled

with the needs of the patient to optimize outcome.

8. Physicians should make an effort to learn more

about the products that have been carefully and

stringently tested.

12 Immune Deficiency Foundation: Clinical Focus October 2003

Page 13: Editor ISSUE 11 OCTOBER 2003 Howard Lederman, M.D. Ph.D ... › sites › default › files › ... · have defective humoral immunity with normal cell-mediated immune function,or

October 2003 Immune Deficiency Foundation: Clinical Focus 13

Page 14: Editor ISSUE 11 OCTOBER 2003 Howard Lederman, M.D. Ph.D ... › sites › default › files › ... · have defective humoral immunity with normal cell-mediated immune function,or

14 Immune Deficiency Foundation: Clinical Focus October 2003

Page 15: Editor ISSUE 11 OCTOBER 2003 Howard Lederman, M.D. Ph.D ... › sites › default › files › ... · have defective humoral immunity with normal cell-mediated immune function,or

15 Immune Deficiency Foundation: Clinical Focus October 2003

1. Fischer A. Primary immunodeficiency diseases:An experimentalmodel for molecular medicine. Lancet 357:1863-1869, 2001.

2. Stuckey M, Quinn PA and Gelfand EW. Identification of ureaplasma urealyticum (T-stain mycoplasma) in a patient withpolyarthritis. Lancet 2:917-920, 1978.

3. Roifman CM,Rao CP,Lederman HM,Lavi S,Quinn P and Gelfand EW.Increased susceptibility to mycoplasma infection in patients withhypogammaglobulinemia . Amer J Med 80:590-594, 1986.

4. Gelfand EW. Intravenous gammaglobulin therapy in immunocompromised patients. In: Intravenous GammaglobulinTherapy, RJ Garner and RA Sacher, eds. American Assoc, BloodBanks (Arlington,VA) pp. 31-46, 1988.

5. Mohiuddin AA, Corren J, Harbeck RJ,Teague JL,Volz M andGelfand EW. Ureaplasma urealyticum chronic osteomyelitis in apatient with hypogammaglobulinemia. J Allergy Clin Immunol87:104-107, 1991.

6. Roifman CM, Levison H and Gelfand EW. High-dose versus low-dose intravenous immunoglobulin in hypogammaglobulinaemiaand chronic lung disease. Lancet I:1075-1077, 1987.

7. Quartier P, Debre M, De Blic J, et al. Early and prolonged intravenous immunoglobulin replacement therapy in childhoodagammaglobulinemia:A retrospective survey of 31 patients.J. Ped. 134:589-596, 1999.

8. Gelfand EW, Reid B and Roifman CM. Intravenous immune serumglobulin replacement in hypogammaglobulinemia: A comparisonof high versus low dose therapy. In: Monographs in Allergy,Vol. 23, pp. 177-186, 1988.

9. Kainulainen L,Varpula M, Liippo K, Svedstrom E, Nikoskelainen J,and Ruuskanen O. Pulmonary abnormalities in patients with primary hypogammaglobulinemia. J.Allergy Clin. Immunol.104:1031-1036, 1999.

10. Busse PJ, Razvi S, and Cunningham-Rundles C. Efficacy of intravenous immunoglobulin in the prevention of pneumonia inpatients with common variable immunodeficiency. J.Allergy Clin.Immunol. 109:1001-1004, 2002.

11. Waniewski J, Gardulf A, and Hammarstrom L. Bioavailability of g-globulin after subcutaneous infusions in patients with commonvariable immunodeficiency. J. Clin. Immunol. 14:90-97, 1994.

12. Eijkhout HW, van der Meer JWM, Kallenberg CGM,Weening RS,van Dissel JT, Sanders LAM, Strengers RFW, Nienhuis H, andSchellekens PTA. The effect of two different dosages of intravenous immunoglobulin on the incidence of recurrent infections in patients with primary hypogammaglobulinemia.Ann. Intern. Med. 135:165-174, 2001.

13. Kazatchkine MD, and Kaveri SV. Advances in immunology:Immunomodulation of autoimmune and inflammatory diseases withintravenous immune globulin. New Eng. J.Med.345:747-755,2001.

14. World Health Organization. Appropriate use of humanimmunoglobulin in clinical practice: Memorandum from anIUIS/WHO meeting. Bull WHO 60:43-47, 1982.

15. Hetherington SV, and Giebink GS. Opsonic activity ofimmunoglobulin prepared for intravenous use. J. Lab. Clin. Med.104:977-986, 1984.

16. Schroeder DD,Tankersley DL, and Lundblad JL. A new preparationof modified immune serum globulin (human) suitable for intravenousadministration. II. Functional characterization. Vox. Sang.40:383-394, 1981.

17. van Furth R, Leijh, PC, and Klein F. Correlation betweenopsonic activity for various microorganisms and composition ofgammaglobulin preparations for intravenous use. J. Infect. Dis.149:511-517, 1984.

18. van Furth R, Braat AG, Leijh PC, and Gardi A. Opsonic activity andphysiochemical characteristics of intravenous immunoglobulinpreparations. Vox. Sang. 53:70-75, 1987.

19. Givner, LB. Human immunoglobulins for intravenous use:Comparison of available preparations for group B streptococcalantibody levels, opsonic activity and efficacy in animal models.Pediatrics 86:955-962, 1990.

20. Steele RW,Augustine RA,Tannenbaum,AS, and Charlton RK.A comparison of native and modified intravenous immunoglobulinfor the management of hypogammaglobulinemia. Amer. J. Med.Sci. 293:69-74, 1987.

21. Darling AJ. Considerations in performing virus spiking experimentsand process validation studies. Dev. Biol. Stand. 81:211-229, 1993.

22. Korneyeva M, Hotta J, Lebing W, Rosenthal RS, Franks L, andPetteway SR, Jr. Enveloped virus inactivation by caprylate:A robust alternative to solvent-detergent treatment.Biologicals 30:153-162, 2002.

23. National Institutes of Health Consensus Development Conference.Intravenous immune globulin:Prevention and treatment of disease.Consensus statement, May 21-23, 1990.Vol. 8, No. 5. Bethesda MD:Department of Health and Human Services, 1990.

24. Renal insufficiency and failure associated with immune globulinintravenous therapy. MMWR 48:1985-1998, 1999.

25. Gaines AR. Acute onset hemoglobulinemia and/or hemoglobinuriaand sequelae following Rho(D) immune globulin intravenousadministration in immune thrombocytopenic purpura patients.Blood 95:2523-2529, 2000.

26. Abzug MJ, Keyserling HL, Lee ML, Levin MJ, and Rotbart HA.Neonatal enterovirus infection:Virology, serology, and effects ofintravenous immune globulin. Clin. Infect.Dis. 20:1201-1206,1995.

27. Fischer GW, Hemming VG, Hunter KW Jr., et al. Intravenousimmunoglobulin in the treatment of neonatal sepsis:Therapeuticstrategies and laboratory studies. Ped. Infect.Dis. J. 5:171-175,1986.

28. Stangel M,Kiefer R,Pette M,Smolka MN,Marx P,Gold R. Side effectsof intravenous immunoglobulins in neurological autoimmune disorders.A prospective study. J Neurol, 250(7): 818-21, 2003.

29. van der Meche FGA, Schmitz PIM.A randomized trial comparingintravenous immune globulin and plasma exchange in Guillain-Barre syndrome. Dutch Guillain-Barre Study Group. N Engl J Med.326(17): 1123-9, 1992.

30. Schiff RI, Sedlack D, and Buckley RH. Rapid infusion ofSandoglobulin in patients with primary humoral immunodeficiency.J.Allergy Clin. Immunol. 88:61-67, 1991.

31. Grillo JA, Gorson KC, Ropper AH, Lewis J, and Weinstein R.Rapid infusion of intravenous immune globulin in patients withneuromuscular disorders. Neurology 57:1699-1701, 2001.

32. Roifman CM, Schroeder H, Berger M, Sorensen R, Ballow M,Buckley RH, Gewurz A, Korenblat P, Sussman G, Lemm G, and theIGIV-C in PIDD Study Group. Comparison of the efficacy of IGIV-C, 10% (caprylate/chromatography) and IGIV-SD, 10% asreplacement therapy in primary immune deficiency. A randomizeddouble-blind trial. Intl. Immunopharm. 3:1325-1333, 2003.

References

Page 16: Editor ISSUE 11 OCTOBER 2003 Howard Lederman, M.D. Ph.D ... › sites › default › files › ... · have defective humoral immunity with normal cell-mediated immune function,or

October 2003 Immune Deficiency Foundation: Clinical Focus 16

what ’s in IGIV?

nex

t

www.gamunex.comwww.IGIVnext .com

Are a l l IGIVs

wel l to lerated?

A review of medical literature

reveals a wide range of IGIV

(Immune Globulin Intravenous

(Human)) related side

reactions.

IGIVs are as unique as the

patients whose lives depend

on them.

At Bayer we believe in the

discovery of new options that

address the needs of patients.

Tolerability can affect

Quality of Life.

Cre

ativ

e by

Eur

o RS

CG

Life

Tor

onto

Page 17: Editor ISSUE 11 OCTOBER 2003 Howard Lederman, M.D. Ph.D ... › sites › default › files › ... · have defective humoral immunity with normal cell-mediated immune function,or

17 Immune Deficiency Foundation: Clinical Focus October 2003

How much t ime should IGIV therapy take from pat ients ’ l ives?

Because all IGIVs (Immune

Globulin Intravenous (Human))

are different, products can vary

in terms of concentration and

infusion time. Historically,

patients had to choose

between:

• speed of delivery or

• tolerability

At Bayer we don’t think patients

should have to make this

trade-off.

Bayer believes in finding

Better Solutions.

Cre

ativ

e by

Eur

o RS

CG

Life

Tor

onto

what ’s in IGIV?

nex

t

www.gamunex.comwww.IGIVnext .com

Page 18: Editor ISSUE 11 OCTOBER 2003 Howard Lederman, M.D. Ph.D ... › sites › default › files › ... · have defective humoral immunity with normal cell-mediated immune function,or

Non-profit Org.U.S. Postage

PAIDAnnapolis, MDPermit No. 273

40 West Chesapeake Avenue, Suite 308 Towson, MD 21204

Address Correction Requested

The Immune Deficiency Foundation(IDF) is the national non-profit healthorganization dedicated to improvingthe diagnosis and treatment of primary immune deficiency diseasesthrough research, education and advocacy. IDF was established morethan two decades ago by concernedfamilies of patients and their physicians. Since its inception, IDF hasexpanded to offer medical education,fellowship and research opportunitiesand publications. IDF sponsors a biennial National Conference forpatients, their families and healthcareprofessionals.

More information about primaryimmune deficiency diseases and IDF can be found at www.primaryimmune.org or by calling 1-800-296-4433.