Chapter 49
Immunology of Isolated and Recurrent Spontaneous Pregnancy Loss
Danny J. Schust and Joseph A. Hill
Main Menu   Table Of Contents

Search

Danny J. Schust, MD
Assistant Professor, Department of Obstetrics/Gynecology and Reproductive Biology, Brigham and Women's Hospital, Boston, Massachusetts (Vol 5, Chap 49)

Joseph A. Hill, MD
Associate Professor of Obstetrics, Gynecology and Reproductive Biology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts (Vol 5, Chap 49)

 
INTRODUCTION
IMMUNOLOGIC PHENOMENA
BASIC IMMUNOLOGIC CONCEPTS
IMMUNOLOGIC INTERACTIONS AT THE MATERNAL—FETAL INTERFACE
IMMUNE CELLS POPULATING THE FEMALE REPRODUCTIVE TRACT
IMMUNE TOLERANCE
IMMUNOLOGIC TESTING
THERAPY
CONCLUSIONS
REFERENCES

INTRODUCTION

Spontaneous pregnancy loss is the most common complication of pregnancy: approximately 70% of human conceptions fail to achieve viability, and an estimated 50% are lost before the first missed menstrual period.1 Recurrent pregnancy loss (RPL) or recurrent spontaneous abortion is much less common, occurring in about 1 in 100 pregnant women.2

Historically, recurrent abortion was defined as three or more clinically recognized pregnancy losses before 20 weeks of gestation. Using this definition, RPL occurs in approximately 1 in 300 pregnancies.3 However, many recommend that clinical investigation and intervention be initiated after two consecutive spontaneous abortions, especially if any of the following are coexistent: fetal heart activity was identified before any of the pregnancy losses, fetal karyotyping of pregnancy tissues revealed normal chromosome content, the woman is older than 35 years of age, or the couple also shows subfertility.

The etiologies of RPL and their respective prevalences are controversial. The only undisputed causes of RPL are parental chromosomal abnormalities and thrombotic complications of the antiphospholipid antibody syndrome (APAS). Most investigators consider the following conditions to be associated with RPL: anatomic abnormalities (congenital or acquired); endocrine abnormalities; infections; and immunologic factors, including those associated with APAS.4,5 Other factors, such as environmental exposures, have been implicated but probably account for less than 10% of cases. The exact proportion of patients affected by each of these factors differs depending on the population being studied. Etiologic proportions may also be affected by referral bias and therefore may differ based on the interests of the investigator conducting a particular study.

After a thorough evaluation, the potential cause for RPL remains unexplained in approximately half of patients.4,5 Investigators have frequently suggested that many of these otherwise unexplained RPL cases may have an immunologic component. Many cases of either isolated or recurrent spontaneous pregnancy loss may have more than one potential etiology, and immunologic mechanisms have been frequently invoked as contributory. This is certainly proposed when infection is associated with pregnancy loss.

Reproductive tract infections with bacterial, viral, parasitic, zoonotic, and fungal organisms have been theoretically linked to both isolated and recurrent pregnancy loss. However, the etiologic mechanisms linking specific organisms to either isolated or recurrent pregnancy loss remain unclear and must certainly differ among infectious organisms.6,7,8 For instance, some viral organisms, such as herpes simplex virus9 and human cytomegalovirus,10 have been shown to infect the placenta/fetus directly, and pregnancy loss related to these infections may be the result of direct damage to the developing fetus by the infectious organism. Alternatively, pregnancy disruption might be an untoward effect of the immune response to the infectious agent. This mechanism may be responsible for some adverse infection-associated events later in gestation, such as intrauterine growth restriction,11 premature rupture of membranes, and preterm parturition.12 Conversely, there is evidence that some of the same mechanisms that potentially protect the fetus from autoimmune rejection may also protect virally infected placental cells from recognition and clearance. Some of the fetoprotective immunoregulatory events that occur at the maternal—fetal interface could thereby make the implanting fetus particularly vulnerable to certain infectious agents, including herpes, cytomegalovirus, and HIV.13

Back to Top
IMMUNOLOGIC PHENOMENA

The number of investigations involving reproductive immunology and its relationship to both isolated and recurrent pregnancy loss has expanded exponentially during the past few decades. Many of these studies, however, have been poorly designed or poorly controlled or have involved insufficient numbers of patients to warrant their conclusions. The clinical study of the immunology of early pregnancy is also hampered by a variety of intrinsic factors. First, a diversity of distinct immune alterations may result in isolated or recurrent pregnancy loss.14 Each of these may act alone or in combination with additional immune or nonimmune factors to result in pregnancy loss. Second, many RPL patients present after their current pregnancy has expired but before its expulsion. Diagnosis of an immunologic cause for that index pregnancy loss is therefore difficult because the expected physiologic immune reaction to the presence of nonviable tissue may mask any alternative underlying immune causes for the demise itself. Prior studies are also compromised because of their failure to correct for embryo and fetal chromosomal abnormalities, which are independent causes of pregnancy loss. Perhaps the most important intrinsic factor is the reassuring fact that for most patients with RPL, the possibility that their next pregnancy will result in a term delivery is quite high. Therefore, very large studies are needed to detect therapeutic effects, including those related to immunomodulatory interventions.

Back to Top
BASIC IMMUNOLOGIC CONCEPTS

A few basic immunologic concepts should be re-examined before a more directed discussion of the involvement of the immune system in pregnancy maintenance.15 Those concepts include the following immunologic subcategorizations: innate versus acquired immune responses; cellular versus humoral immune responses; antigen presentation by major histocompatibility complex (MHC) class I versus MHC class II molecules; and peripheral versus mucosal immune systems.

Each of these subdivisions is related to the others, and each is useful in understanding particular aspects of investigations on the immune interactions at the maternal—fetal interface.

Innate Versus Acquired Immune Responses

Immune responses are classically divided into innate and acquired. Innate responses represent the body's first line of defense against pathogenic invasion. Innate responses are rapid and are not antigen-specific. Cell types and mechanisms typically considered vital to innate immunity include complement activation, phagocytosis by macrophages, and lysis by natural killer (NK) cells and possibly by T cells expressing the T-cell receptor (TCR)-γδ (see below). Acquired immune responses, in contrast, are antigen-specific and are largely mediated by T and B cells. Acquired responses can be classified as either primary (a response associated with initial antigen contact) or secondary (a rapid and powerful amnestic response associated with repeated contact to the same antigen).

Cellular Versus Humoral Immune Responses

Further subdivision of acquired immunity into either cellular or humoral responses is useful, even though it grossly oversimplifies the complexity of immune interactions. By definition, the ability to mount a cellular immune response to a particular antigen can be transferred to a naïve (nonimmunized) individual via the lymphocytes (but not plasma or serum) from another immunized subject. Conversely, humoral immune responsiveness to a particular antigen can be transferred to a naïve subject using only the plasma or serum from an immunized individual. No cells need be transferred, and the response is known to be dependent on the presence of antibodies in the immunizing sera. Put simply, cellular responses require cell-to-cell interactions, whereas humoral responses are antibody-mediated. Like innate and acquired immunity, these cellular and humoral responses are intricately intertwined.

Antigen Presentation by MHC Class I Versus MHC Class II Molecules

Cellular and humoral immune responses are largely dependent on the presence of two sets of genes in the MHC that play major roles in determining antigen specificity. These genetic loci encode the MHC class I and MHC class II products, as well as many of the supporting effector molecules involved in antigen presentation. Both MHC class I and MHC class II molecules help to alert the immune system to alterations that would require an immune response. MHC class I molecules (HLA-A, -B, and -C) are present on the surface of nearly every cell in the human body and are important in defense against intracellular pathogens, such as viruses, and against oncogenic transformation. MHC class I molecules act as important ligands for the T-cell receptor on CD8+ cytotoxic/suppressor T cells and for a variety of receptors on NK cells.16 MHC class I-mediated intercellular interactions generally activate the cellular portion of the immune response and result in killing of the antigen-presenting (i.e., virally infected or cancerous) cell expressing the class I molecule.

In contrast, MHC class II molecules (HLA-DR, HLA-DP, and HLA-DQ) are present on the surface of a limited number of “professional” antigen-presenting cells. These include dendritic cells, macrophages, monocytes, B cells, and tissue-specific antigen presenters (e.g., Langerhans cells in the skin). MHC class II molecules are important in defense against extracellular pathogens, such as bacterial invaders. The major ligand for MHC class II is the T cell receptor on CD4+ T-helper cells. MHC class II-mediated interactions generally result in the modulation of humoral immune responses.

Peripheral Versus Mucosal Immune Systems

Perhaps the most relevant immunologic categorization to the discussion of pregnancy maintenance is that which divides the immune system into two distinct compartments: peripheral and mucosal. In general, basic immunologic principles have been tested and described for the immune effector cells populating only one of these compartments—the peripheral immune system. Until recently, very little was known about the mucosal immune compartment, and its description continues to lag far behind that of the periphery.

The peripheral immune system consists of the spleen and peripheral blood, and it is generally responsible for protection against blood-borne pathogens. In contrast, the mucosal immune system is typically the first to encounter those pathogens that enter via the extensive surface areas of the lacrimal ducts, respiratory and gastrointestinal tracts, mammary ducts, and genitourinary tracts. Thus, the mucosal immune system appears to be primarily responsible for providing at least initial immunologic protection against the majority of exogenous pathogens.

Back to Top
IMMUNOLOGIC INTERACTIONS AT THE MATERNAL—FETAL INTERFACE

Most of what we now know about the specific characteristics of immune interactions at mucosal sites arose during the past two decades. The first of the mucosal immune sites to be extensively studied was the gastrointestinal tract, and investigations revealed its immunology to be quite distinct from that of the periphery.17 Studies extending to the reproductive tract documented immunologic distinctions from both the periphery and the gastrointestinal tract.18 Further application to immunologic interactions at the site of implantation have revealed additional unique immunologic characteristics. For instance, descriptions of the immune cells that populate both the female reproductive tract and the peri-implantation decidua implicate a particularly important role for innate immune interactions during implantation and early pregnancy.19,20

Back to Top
IMMUNE CELLS POPULATING THE FEMALE REPRODUCTIVE TRACT

Investigations on the immune cells of the female reproductive tract, particularly those populating the decidua at the time of implantation and during early pregnancy, have identified four potentially important lines of investigation based on four unusual immune cell types: decidual granular lymphocytes, NKT cells, TCRγδ+ cells, and supressor macrophages.

Decidual Granular Lymphocytes

Whereas the human endometrium is normally populated by T cells, macrophages, NK-like cells, and a very limited number of B cells,19,20,21 the relative proportions of these resident cells show menstrual cyclicity. The most dramatic changes are noted during the late luteal phase and in early pregnancy, when the proportion of one unusual type of immune cell rises to nearly 70% to 90% of the total endometrial lymphocyte populations.19,20,21 These unusual cells have been variably called decidual granular lymphocytes (DGLs), large granular lymphocytes (LGLs), and decidual NK cells. The origin, functional capabilities, and physiologic purpose of these cells remain enigmatic, but their abundance at sites of implantation compels further study.22,23 Although this particular cell type differs from similar cells isolated from the periphery, most believe it to be an NK cell variant. If so, the implantation site represents the largest accumulation of NK cells in any state of human health or disease.

NKT Cells

Peripheral immune cells have been described that have characteristics of both NK cells and T cells.24 These NKT cells and their ligands (e.g., CD1) have recently been shown to be present in the decidua of animals25,26,27 and have been implicated in some forms of pregnancy loss.25 In addition, the presence of NKT cells at the implantation sites of murine pregnancies has been shown to be mediated by interactions with fetally expressed MHC class I or class I-like products.26 The presence of NKT cells and their ligands in human decidua is under investigation.

TCR γ δ+ Cells

In the peripheral immune compartment, the vast majority of T cells express a T-cell receptor composed of an αβ heterodimer (TCRαβ+). In addition to TCRαβ+ T cells, the human reproductive tract is also populated by a subset of T cells with a distinctive T-cell receptor, the γδ heterodimer (TCRγδ+ cells). The number of these cells increases in early pregnancy.28,29,30 TCRγδ+ T cells appear to fulfill functions quite distinct from their TCRαβ+ counterparts; these functions may include direct, non-MHC-restricted recognition of antigens within tissues.31 TCRγδ+ T cells may fill a protective niche missed or poorly addressed by B cells and TCRαβ+ T cells. The role and importance of TCRγδ+ cells in the reproductive tract and in pregnancy maintenance needs further study.

Suppressor Macrophages

A subset of macrophages termed suppressor macrophages may be involved in pregnancy maintenance. These specific cells differ from typical macrophages in their promotion of anti-inflammatory effects and they have been detected in normal murine placenta.32 Their presence in human decidual specimens has not been definitively investigated.

In conclusion, very characteristic immune effector cells populate the human decidua. To date, insufficient patient numbers have hampered investigations into whether alterations in these cellular populations (including T cells, decidual NK cells, and NKT cells) determine pregnancy outcome. Still, the general consensus of these studies is that these populations are altered in RPL patients33,34,35,36,37 but not in patients experiencing isolated spontaneous pregnancy losses.30

Back to Top
IMMUNE TOLERANCE

Immune Cell Education and Homing to the Female Reproductive Tract

One very important concept in immunology that has particular application to pregnancy is that of immune tolerance. The concept is very well described for the immune cells of the peripheral immune system.15 Those immune cells arising in the bone marrow and destined to become T lymphocytes typically pass through the fetal thymus, where they undergo a process termed thymic education. During thymic education, T cells “mature” from CD4/CD8 double-positive cells to single-positive (CD4+ or CD8+) cells. This maturation occurs by a two-step process that spares only those T cells capable of recognizing one's own MHC class I or class II molecules but incapable of recognizing other self peptides. This education promotes T-cell self-tolerance by allowing selection and survival only of T cells that will not react against the organism's own peptides.

If one considers the implanting blastocyst as the most common example of a successful allograft, one must question how the concepts of tolerance apply at the maternal—fetal interface against tissue that is most certainly not self. A few concepts require attention. First, are there characteristics other than those phenotypic differences already described that promote tolerance to fetal antigens among the resident immune cells in the maternal decidua? Both animal and human data suggest that the immune cells at the maternal—fetal interface are selected and maintained in ways that differ both from peripheral immune cells and from cells populating other mucosal sites, such as the intestine.18,38 Second, if the cells populating the maternal—fetal interface have distinctive phenotypes and antifetal reactivity, how are these particular cells recruited to the site of implantation? Cellular recruitment (homing) to the intestine has been shown to rely on the presence of receptors (integrins) on the surface of particular immune cell subpopulations that specifically interact with ligands on the surface of the endothelial cells of blood vessels within the intestine (e.g., selectins, VCAM, MECAs).39,40 Similar homing characteristics also appear to define immune cell types and vascular structures within the reproductive tract.39,40,41,42

Control of Decidual Immune Cell Immunoreactivity

Because safeguarding the fetus from maternal immune rejection is obviously crucial for successful reproduction, it is not surprising that evolution has selected a wide variety of somewhat redundant immune adaptations at the maternal—fetal interface. In addition to distinctions in phenotype, homing, and antigenic education, the immune cells that populate the maternal decidua also appear to be affected by local immunomodulation. We know from both human and animal studies that immune responses to fetal antigens can be detected.43,44,45 Thus, the regulation of these responses at the maternal—fetal interface may be critical for pregnancy maintenance. The concept that successful pregnancy requires some form of suppression of the maternal immune response is supported by reports that failure to downregulate maternal responses to recall antigens, such as tetanus toxoid and influenza, is associated with poor pregnancy outcome among RPL patients.46 Some of the mechanisms hypothesized to be important in this localized immune regulation include alterations in localized cytokine profiles, the immune effects of pregnancy-associated hormones, and the in situ tolerance-promoting characteristics of tryptophan metabolism.

CYTOKINE DYSREGULATION.

An essential role for isolated cytokines in reproductive success or failure is not well supported by animal studies. To date, although most cytokines have been gene-deleted in separate animal models, only one of these factors appears to be essential to pregnancy maintenance: leukemia inhibitory factor is required for blastocyst implantation47 but not for subsequent embryogenesis.48 Although this does not argue that other cytokines and soluble immunoregulatory factors are inconsequential, there certainly seems to be significant redundancy among these substances at the site of implantation.

It may not be particularly relevant to examine these factors in isolation. Rather, groups of cytokines may best reflect immune responses.49,50 The division of antigen-stimulated immune responses involving CD4+ T cells into T helper 1 (Th1) responses and T helper 2 (Th2) responses appears to be useful, albeit somewhat oversimplified. This subclassification is defined both by the characteristics of the CD4+ cells present and by the cytokines these cells secrete. In general, the characteristics of the cytokine response depends on the environment in which naïve CD4+ (Th0) cells mature and differentiate: Th0 cells exposed to interferon (IFN)-γ become Th1-type cells, and those exposed to interleukin (IL)-4 become Th2-type cells.49,50 Th1 responses are associated with inflammation and are typically characterized by the release of IFN-γ, IL-12, IL-2, and tumor necrosis factor (TNF)-β. Th2 cell responses are associated with antibody production and the cytokines IL-10, IL-4, IL-5, and IL-6.51,52,53 Although TNF-α can be secreted by both Th1 and Th2 cells, it is most often associated with a Th1 response.54,55 Th1 and Th2 responses are intimately interdigitated, with each type of response promoting itself while limiting the alternative response.56,57,58,59

The human endometrium and decidua are replete with immune and inflammatory cells capable of cytokine secretion.60,61,62,63,64 Extension of the Th1/Th2 paradigm to pregnancy has generated provocative hypotheses. Most women with normal pregnancies appear to have a predominant Th2 immune response to undefined trophoblast antigens.65 In contrast, some patients with recurrent pregnancy loss exhibit a dysregulation of their T-helper response to antigens at the site of implantation, with shifts toward Th1 inflammatory responses.65,66,67,68 Further, Th1-type cytokines have been shown to be harmful to an implanting embryo.69,70,71 Depending on the individual series, 15% to 20% of nonpregnant women with a history of otherwise unexplained recurrent spontaneous abortion have been found to have evidence of abnormal in vitro Th1 cellular immune responses to trophoblast antigens. Fewer than 3% of women with normal reproductive histories have these responses.65,66,68,72

The literature concerning reproductive immunology is often frustratingly inconsistent. Part of the frustration that arises when one attempts to synthesize the data surrounding cytokine dysregulation and poor reproductive outcome lies in the variations among study designs. Methods for the documentation of cytokine dysregulation among RPL patients have included confirmation of the abnormality within the endometrium73,74,75 and the decidua.76 Other investigators use peripheral lymphocytes from women with a history of RPL and stimulate them in vitro with trophoblast antigens.65,77,78 Whether peripheral cytokine levels reflect T-helper cell dysregulation at the maternal—fetal interface and whether this localized dysregulation affects peripheral as well as local immune responses during pregnancy remain controversial.79,80,81

REPRODUCTIVE HORMONES.

Notable gender differences in immune responsiveness82 may reflect the fact that reproductive hormones have dramatic effects on peripheral cell-mediated immunity. Both estrogen and progesterone have been shown to be potent immunomodulators, although the latter has received the most attention in terms of implications for the maintenance of the semiallogeneic implanting conceptus.83 Recent in vitro evidence supports progesterone's immunosuppressive attributes by demonstrating an inhibition of mitogen-induced proliferation of and cytokine secretion by CD8+ T cells.84 Further, progesterone may promote the development of a cytokine microenvironment favoring pregnancy maintenance. Progesterone-mediated changes in T-cell gene expression have been associated with the Th2-type T-helper cell responses and with increased expression of leukemia inhibitory factor.76,85 Because a shift in the intrauterine immune environment from Th2 to Th1 has been linked with early pregnancy loss,65,76 the elevated concentrations of progesterone characteristic of early pregnancy may promote an immune environment favoring pregnancy maintenance.

Although not specifically addressing pregnancy, several reports have recently focused attention on the immunomodulatory effects of estrogens. Estrogens have been shown to improve immune responses in men after significant trauma/hemorrhage,86 to suppress cell-mediated immunity after thermal injury,87 and to protect against chronic renal allograft rejection,88 all in animal models. In vitro, estrogens downregulate delayed-type hypersensitivity reactions and promote the development of Th2-type immune responses, particularly when present in high, and often supraphysiologic, concentrations.89,90,91

Of course, one obvious question poses itself when hypotheses on progesterone/estrogen immunomodulation and their role in pregnancy maintenance are examined. If elevated levels of progesterone and estrogen are sufficient to promote tolerance to the implanting fetus, are pregnant women also systemically immunosuppressed? The answer to this question is complex. Some forms of immunoreactivity are indeed systemically suppressed in pregnant women. Examples of these effects include the symptomatic remission of such autoimmune disorders as rheumatoid arthritis among pregnant women with disease resurgence postpartum.92 Pregnancy also appears to confer protective effects on the progression of multiple sclerosis.93 Further, some viral diseases, including varicella, are particularly aggressive when first encountered during pregnancy.94 In addition, although the levels of potentially immunosuppressive hormones are systemically elevated in pregnant women, their concentrations at the maternal—fetal interface appear to be significantly higher than in the maternal circulation.95

TRYPTOPHAN METABOLISM.

A novel immunoregulatory pathway involving the amino acid tryptophan and its catabolizing enzyme, indoleamine 2,3 dioxygenase (IDO), has recently been proposed as a mechanism for maternal tolerance to the fetal allograft. T cells require tryptophan for activation and proliferation.96,97 Local alterations in tryptophan supply or in tryptophan metabolism at the maternal—fetal interface would therefore be predicted to modulate immune cell function at this site. Evidence implicating tryptophan metabolism in maternal—fetal immune interactions arose initially in animal models. The inhibition of IDO in mice has been shown to promote loss of allogeneic but not syngeneic fetuses, an effect mediated by lymphocytes.98 Further, hamsters fed diets high in tryptophan have increased rates of fetal wastage.99 IDO has been reported to be expressed in human uterine decidua.100 Alterations in serum tryptophan levels with increasing gestational age during human pregnancy have also been noted.101

MHC Antigens in the Placenta

Using available reagents and investigative techniques, early investigators failed to detect MHC-encoded transplantation antigens in the placenta. It was already known that these MHC class I and class II antigens promoted the rejection of transplanted allogeneic tissue in nonreproductive systems. Therefore, their proposed absence on the fetal semiallograft fostered the hypothesis that the maternal immune cells were tolerant of an implanting pregnancy because the invading tissues simply were not recognized as foreign. This hypothesis, and the data on which it was based, are partially correct.

MHC class II molecules are not expressed on the surface of placental trophoblast cells,102,103 nor are the classical MHC class I transplantation antigens HLA-A and -B. However, a group of unique MHC class I products—HLA-C, HLA-E, and HLA-G—are expressed on a subpopulation of placental cells called extravillous cytotrophoblast cells.13,104,105,106,107,108,109 Extravillous cytotrophoblast cells are derived from the villous core of the placental cotyledon. During the course of early placental maturation, these cells take on invasive qualities. Invasion of extravillous cytotrophoblast cells may be regulated by a number of factors, including MHC expression patterns, integrin expression patterns/integrin switching,110 and in situ oxygen tension. Invasion is characterized initially by movement of the extravillous cytotrophoblast cells from the tips of the placental anchoring villae toward positions deep within the maternal decidua. These cells then continue their invasion, reaching the maternal decidual vasculature, and ultimately replace cells within the walls of arterial decidual vessels.111,112 At this site, these fetally derived extravillous cytotrophoblast cells intimately contact maternal immune effector cells, thereby exposing the fetus to potential MHC-restricted recognition as nonself.

The MHC class I molecules on the extravillous cytotrophoblast cell are represented by the classical MHC class I product HLA-C and the nonclassical HLA-E and HLA-G products. Several lines of investigation have been pursued to determine why all placental cells downregulate expression of the classical transplantation antigens (HLA-A and HLA-B), whereas invasive extravillous cytotrophoblasts express HLA-C, HLA-E, and HLA-G.13,19,20,22,97 There may be functional similarities among HLA-C, HLA-E, and HLA-G that promote recognition by immune cells at the maternal—fetal interface and downregulation of antifetal responses. Interactions of these class I products with the abundant NK-like immune cells at the maternal—fetal interface may avert NK cell receptor-mediated killing of extravillous cytotrophoblast. Interactions of trophoblast HLA-C, HLA-E, and/or HLA-G with surface receptors on either NK cells or other maternal immune effector cells may modulate immune cell cytokine expression profiles.22,23 Supporting the latter hypothesis, aberrant cytokine secretion has been documented when peripheral blood lymphocytes from RPL patients are stimulated by HLA-G-bearing cells in vitro.113 Further, both decidual and peripheral immune cells have been shown to shift toward the Th2 phenotype when exposed to HLA-G.114 The expression by fetus-derived cells of any or all of the trophoblast MHC class I products could also promote essential decidual and vascular invasion.115 Altered trophoblast expression of HLA-G has been linked to disorders of placental invasion, such as pre-eclampsia.115,116 Finally, the effects of MHC class I products on maternal reactivity toward the implanting pregnancy could be indirect. Secretion of soluble forms of HLA-C, HLA-E, and/or HLA-G into the maternal circulation may have protolerance immune inductive effects at sites distant from the maternal—fetal interface.117

Although associations between HLA typing and recurrent pregnancy loss have been suggested (see below), very few investigations have specifically addressed the role of trophoblast MHC class I products (HLA-C, HLA-E, and HLA-G) in pregnancy loss. Two small studies investigating a possible association between polymorphisms in these MHC genes and the occurrence of RPL have been negative.118,119 Others have addressed the possibility that because trophoblast cells do not spontaneously express either classical HLA-A and HLA-B products nor MHC class II products, an aberrant expression of these molecules might result in an adverse maternal immune response to the implanting fetus. Data supporting this hypothesis remain limited.120,121,122

Humoral Immune Mechanisms

The literature is controversial concerning the study of humoral/antibody-mediated immune responses and isolated or recurrent spontaneous pregnancy loss. In fact, much of this literature has now become obsolete. Responses to pregnancy-specific antigens exist, and patients with RPL can display altered humoral responses to endometrial antigens,123 but investigators continue to debate whether recognition of fetal antigens is essential/protective or harmful.

IMMUNOPROTECTION: BLOCKING ANTIBODY DEFICIENCY.

One hypothesis promoting maternal recognition of the implanting pregnancy as essential rests on the supposition that humoral immune-mediated pregnancy loss results from a deficiency in the maternal production of protective (blocking) factors. The production of these protective factors (presumably antibodies) was thought to be the result of immune recognition of fetus-derived (paternal) alloantigens. It was suggested that these protective factors, termed “blocking antibodies,” would prevent the maternal, cell-mediated, antifetal immune response that was believed to occur in all pregnancies. The production of “blocking antibodies” was therefore deemed crucial so that, in the absence of these factors, abortion would occur.124

A deficiency in “blocking antibodies” was demonstrated in vitro by maternal hyporesponsiveness in mixed lymphocyte culture with paternal stimulator cells.124 Hyporesponsiveness in these assays was associated with RPL;45,125,126 however, this association is now thought to represent the immune effects of repeated losses rather than the cause of pregnancy loss. Further evidence refuting the blocking antibody hypothesis in immune-mediated pregnancy loss comes from reports of successful pregnancies among women who do not produce serum factors capable of mixed lymphocyte culture inhibition124 and among women who do not produce antipaternal cytotoxic antibodies.125

TROPHOBLAST-LYMPHOCYTE CROSS-REACTIVE ANTIGENS.

Out of the blocking antibody theory and its associated investigations came a number of related propositions that are no longer invoked. One of these involved the description of what was thought to be a novel HLA-linked alloantigen system called trophoblast-lymphocyte cross-reactive antigens, or TLX. The presence of TLX was first suggested by reports that polyclonal rabbit antisera could recognize both lymphocytes and trophoblast cells.127 A number of initial reports then linked TLX to maternal blocking antibody deficiency and recurrent pregnancy loss.128,129 TLX, however, was subsequently found to be identical to CD46, a complement receptor that is thought to protect the placenta from complement-mediated attack.130 CD46 is not a novel alloantigen; it is not even an alloantigen. CD46 can be found on a wide variety of cells and has no significant role in distinguishing self from nonself.

HLA SHARING.

Linked to the hypothesis that maternal recognition of the fetal antigens was essential to pregnancy maintenance were studies suggesting that parental HLA (classical MHC class I) sharing predisposed a couple with RPL to blocking antibody deficiency.131,132 According to this theory, if maternal and paternal HLA products were too similar, fetally expressed paternal alloantigens would not elicit a maternal immune response to produce “blocking antibodies.” The two largest studies addressing this possibility used a small religious sect, the Hutterite community, as their study population. The first study was prospective, with population-based controls, and demonstrated that HLA heterogeneity was not essential for successful pregnancy among the Hutterites.133 A follow-up study, however, reported contrasting results. In fact, among the Hutterite population studied in this more recent, 10-year, prospective trial, complete sharing of the entire HLA region was associated with an increase in spontaneous pregnancy loss.134 The conclusions drawn by these authors should be considered carefully, because the incidence of complete sharing of the HLA region between sexual partners in outbred populations is exceedingly rare;only isolated and significantly inbred populations have a chance for such HLA homogeneity. Therefore, the authors conclude that HLA typing is of no clinical utility in outbred populations. A similar link between MHC class II typing and recurrent pregnancy loss has also been reported.135,136

BYSTANDER EFFECT: AUTOIMMUNITY AND PREGNANCY LOSS.

Antibody-mediated mechanisms for recurrent abortion in which humoral immune reactivity might directly result in adverse antifetal effects have been proposed. These include reports on associations between RPL and the presence of antisperm and antitrophoblast antibodies; however, each has been subsequently shown to have minimal clinical relevance.6,121 In contrast, demonstrated associations between pregnancy loss and autoantibodies directed against nonreproductive tissues provide indirect evidence for humoral immune-mediated pregnancy loss. For instance, antithyroid antibodies have been linked to adverse pregnancy outcomes. One large, retrospective study reported an increased prevalence of these antibodies among women with a history of RPL, even in the absence of thyroid endocrinologic abnormalities.137 Others have reported similar findings.138 In contrast, two recent studies addressing pregnancy outcome and the presence of serum antithyroid antibodies have demonstrated neither an association with RPL nor a need for antithyroid antibody testing among RPL patients.139,140

Antiphospholipid Antibodies

More consistent, and more therapeutically relevant, have been reports linking organ-nonspecific autoantibodies associated with APAS to adverse pregnancy outcomes.

Clinical and laboratory evidence for antiphospholipid antibodies were originally based in vivo on thrombosis and in vitro on prolongation in one of the phospholipid-dependent coagulation tests such as the activated partial thromboplastin time or the Russell viper venom time. With time, patterns among the thrombotic complications associated with antiphospholipid antibodies were noted and criteria proposed that defined APAS. Although many of the complications of APAS are systemic, some are pregnancy-specific, including spontaneous abortion, premature labor, premature rupture of membranes, stillbirth, intrauterine growth restriction, and pre-eclampsia.141 The importance of pregnancy-specific complications in the diagnosis of APAS gained renewed prominence when the defining criteria for the disorder were reassessed in 1998 (the Sapporo criteria, Table 1, see below).142,143

Table 1. Sapporo Criteria for Diagnosing the Antiphospholipid Antibody Syndrome

  One or more of the following clinical and one or more laboratory criteria must be present in the same patient:
  Clinical

  1. One or more confirmed episodes of vascular thrombosis of any type, including:
    • Venous
    • Arterial
    • Small vessel

  2. Pregnancy complications, including:
    • Three or more consecutive spontaneous pregnancy losses at less than 10 weeks of gestation
    • One or more fetal deaths at greater than 10 weeks gestation
    • One or more premature births at less than 34 weeks gestation secondary to severe pre-eclampsia or placental insufficiency


  Laboratory
  (testing must be positive on two or more occasions, 6 weeks or more apart)
  1. Positive plasma levels of anticardiolipin antibodies of the IgG or IgM isotype at medium to high levels
  2. Positive plasma levels of lupus anticoagulant

(Adapted from Wilson WA. Gharavi AE. Koike T et al: International consensus statement on preliminary classification criteria for definite antiphospholipid syndrome: Report of an international workshop. Arthritis Rheum 42:1309–1311, 1999.)

Clinical data have linked antiphospholipid antibodies such as anticardiolipin or lupus anticoagulant to adverse pregnancy outcome.144 In a large series of couples with recurrent abortion, the incidence of APAS was 3% to 5%.6 The presence of anticardiolipin antibodies among patients with known systemic lupus erythematosus may be particularly worrisome.145 Pathologic data, however, do not consistently demonstrate causal involvement of APAS in pregnancy loss. For instance, although placental infarction, abruption, and hemorrhage are considered to be defining placental lesions in APAS, they are often missing in women with antiphospholipid antibodies.146 Conversely, women with recurrent abortion but no biochemical evidence of antiphospholipid antibodies often demonstrate placental findings consistent with the diagnosis of APAS.147

Antiphospholipid antibodies have mechanistic links to placental thrombotic changes and subsequent adverse pregnancy outcome. Antiphospholipid antibodies may indirectly promote thrombosis. Associations between antiphospholipid antibodies and alterations in prostacyclin/thromboxane metabolism have been demonstrated. Local alterations in these pathways at the level of the maternal—fetal interface could promote vascular constriction, platelet adhesion, and placental infarction.148,149,150 Similarly, antiphospholipid antibody positivity has been linked to reductions in the levels of placental antithrombotic products, such as annexin V, among women with RPL.151 Antiphospholipid antibodies may promote localized atherosclerosis. To this point, atherosclerosis has been shown to develop rapidly in the spiral arteries of patients positive for antiphospholipid antibodies.152 Antiphospholipid antibodies may impair appropriate placental maturation, differentiation, and invasion. To illustrate, IgM against phosphatidylserine inhibits the formation of syncytial trophoblast153 in cell cultures and sera from antibody-positive RPL patients inhibits trophoblast adhesion to endothelial cells in vitro.154

Attention in the field of antiphospholipid antibodies and pregnancy loss has recently turned partially away from the antigens cardiolipin and phosphatidylserine but toward a protein cofactor called β2 glycoprotein 1.155,156 This glycoprotein acts as an essential cofactor in antiphospholipid antibody reactivity against membrane phospholipid and, in many instances, may provide the true antigenic determinant for a given antiphospholipid antibody.155,156 Because both extravillous cytotrophoblast and syncytiotrophoblast cells have been demonstrated to synthesize β2 glycoprotein 1,157 the placental microenvironment has the potential for robust antigenicity. At present, however, the prognostic value of serum levels of specific antibodies against β2 glycoprotein 1 with respect to pregnancy outcome in RPL patients remains unclear.158,159

Back to Top
IMMUNOLOGIC TESTING

Although much is now known about the immunology of the human genital tract, many questions remain. Immunologic interactions at the maternal—fetal interface are particularly intriguing, but in most cases, basic immunologic findings in the laboratory have yet to be fully applied toward diagnostic or therapeutic use at the bedside. Much progress will undoubtedly be made during the next decade; however, in this era of evidence-based medicine, very few immunologic tests or therapies are presently warranted in the diagnosis and treatment of patients with isolated or recurrent spontaneous pregnancy loss. Most of these tests remain experimental at best and demand further investigation before they can be clinically applied. Still others are of historical interest only.

Antibody Testing

As mentioned above, the 1998 reassessment of the criteria used for the diagnosis of APAS made complications of pregnancy defining clinical criteria.142 A brief review of the Sapporo criteria (see Table 1) demonstrates that although most patients with an isolated spontaneous pregnancy loss do not warrant testing for APAS, many patients with recurrent losses do require further laboratory assessment. Screening for the lupus anticoagulant (activated partial thromboplastin time or Russell viper venom testing) and assessment for the presence of anticardiolipin antibodies is indicated for most patients with RPL but few with isolated losses.

Although investigators have suggested testing for a large number of isolated organ-specific and nonspecific autoantibodies among patients with RPL, testing for antibodies other than the lupus anticoagulant and anticardiolipin antibody remains controversial.160,161 For example, although some have shown the prevalence of the organ-specific antithyroid antibodies to be increased among patients with a history of RPL,137,139 others have found no association between the presence of antithyroid antibodies and recurrent loss.138 Even if the prevalence of antithyroid antibodies is increased among these patients, their clinical and mechanistic significance remains unclear.140

As mentioned above, investigators have demonstrated an association between some organ-nonspecific antiphospholipid antibodies (e.g., antiphosphatidylserine and anti-β2 glycoprotein 1) and abnormal placental maturation, invasion, and development.155,156,157,158,159 However, such correlations do not warrant patient testing. Investigations that clinically link the presence of antiphosphatidylserine and/or anti-β2 glycoprotein 1 to isolated or recurrent pregnancy loss remain limited.162

There is some evidence that a very specific subset of patients—those with known autoimmune diseases and RPL—may warrant some antiphospholipid testing in addition to the lupus anticoagulant and anticardiolipin antibodies.163 There is no consistent evidence supporting the use of extensive panels of serum or site-specific autoantibodies or alloantibodies (including antinuclear antibodies and antipaternal cytotoxic antibodies) in the evaluation of either isolated or recurrent pregnancy loss. Use of these panels often serves only to verify the statistical tenet that if the number of tests performed reaches a critical limit, at least one will be positive in every patient.

Cytokine Testing

The hypothesis that cytokine dysregulation (Th1/Th2 imbalance) at the maternal—fetal interface may affect pregnancy maintenance is gaining momentum. There exists increasing evidence that these effects may be clinically important in some cases of pregnancy loss.19,164 Our ability to diagnose such dysregulation, however, has been hampered by a number of variables, many of which are difficult to circumvent. We have herein described that the immune cells populating the maternal—fetal interface are unique and characteristic. Therefore, although Th1/Th2 dysregulation may be occurring within the immune microenvironment at the site of implantation, peripheral immune cells may likely be spared. Safe methods to assess the immune characteristics at the site of an ongoing and desired pregnancy do not presently exist. Can significant alterations in cytokine profiles that are isolated within the uterus be detected using peripheral serum testing? This remains unclear. Some small studies have documented peripheral shifts toward Th1 cytokine profiles in RPL patients who subsequently lose their pregnancy, but not in similar patients who have a successful pregnancy outcome.81 Other larger studies have failed to show any association between peripheral cytokine alterations and pregnancy outcome among patients with a history of recurrent losses.79 One additional caveat is that pregnancy demise is itself accompanied by dramatic immunologic alterations within the uterus toward the presence of necrotic tissues. Therefore, studies on cytokine dysregulation, including those reporting a peripheral shift toward Th1 profiles at the time of fetal demise among patients with RPL,80 may not be able to differentiate cause versus effect.

Immune Cell Evaluation

Efforts have been made to evaluate both the proportions and activity of immune cells isolated from patients with a history of spontaneous pregnancy loss. These studies are also limited both by the inability to sample sites of implantation during desired pregnancies and by the tenuous association between most decidual and peripheral immune characteristics. Diagnostic use of mixed lymphocyte cultures, originally proposed to evaluate the presence of “blocking antibody” activity, has not been consistently validated. The identification of large numbers of NK-like cells at sites of implantation led to the hypothesis that alterations in the prevalence and activity of these cells might significantly affect pregnancy maintenance. Although the rationale may be questioned, this hypothesis has been extended to promote testing of the prevalence and activity of peripheral NK cells among patients with RPL.165,166 Again, because decidual and peripheral NK cells are markedly different, one must resolve whether intrauterine events are reflected using NK cells isolated from the periphery of these women. Two small studies have nevertheless reported that the evaluation of these peripheral NK cell characteristics predicts prognosis and assists in the counseling of patients with RPL.165,166 These studies have not been substantiated.

HLA Screening

Testing for parental HLA profiles is not indicated in outbred populations. Those reports demonstrating that HLA sharing was associated with poor pregnancy outcomes were strictly limited to the specific populations studied.133,134 In the most recent and definitive of these investigations,134 only patients with complete sharing of the entire HLA locus demonstrated an increased risk for adverse pregnancy outcomes. As discussed, the chance of discovering complete sharing of the entire HLA region among partners in outbred populations is infinitesimally small. Therefore, generalized HLA screening of patients with either isolated or recurrent spontaneous pregnancy loss cannot be rationalized.

Back to Top
THERAPY

Like immunologic testing, “immunologic” therapies for patients with presumed immunologic pregnancy losses have been best substantiated among patients with concomitant hypercoagulability. Treatment regimens for patients with adverse pregnancy outcomes and APAS is appropriately discussed alongside other proposed therapies for immune-mediated pregnancy loss, even though the treatments are themselves best subcategorized as antithrombotic rather than immunomodulating. However, the antithrombotic agent heparin has been shown to bind to antiphospholipid antibodies in vitro, an effect that suggests that it possesses distinct immunoregulatory properties.167

Antithrombotic Therapy

ASPIRIN AND UNFRACTIONATED HEPARIN.

The safety and efficacy of the combined use of aspirin (ASA) and heparin has been best studied among pregnant women who carry the diagnosis of APAS. Although variations have been described,168,169,170 a typical medical regimen for the antithrombotic management of these patients is shown in Table 2.

Table 2. Antithrombotic Treatment of Antiphospholipid Antibody Syndrome During Pregnancy


Aspirin

75–85 mg

Begin with

 

 orally QD

 attempts at

 

 

 conception

Unfractionated sodium

5,000–10,000 IU

Begin with

 heparin

 subcutaneously,

 confirmation

 

 BID

 of pregnancy

Monitoring

Check activated partial thromboplastin time each week; adjust dosage to maintain anticoagulation

Therapy with combinations of ASA and unfractionated heparin in this patient population is not without associated risks. Increases in the incidence of pregnancy-specific complications, including premature labor, premature rupture of the membranes, intrauterine growth restriction, intrauterine fetal demise, pre-eclampsia, and abruptio placenta have all been reported.169,170,171 Specific maternal risks include gastric bleeding and osteopenia. Pregnant patients with APAS should be considered high risk and managed in conjunction with a perinatologist.

LOW-MOLECULAR-WEIGHT HEPARIN.

The inconvenient dosing schedules and unacceptable side effect profiles associated with unfractionated heparin use led to the development of low-molecular-weight heparin (LMWH). When compared with unfractionated heparin, LMWH has an increased antithrombotic ratio, allowing treatment of inappropriate clotting with fewer bleeding side effects.172 The incidence of thrombocytopenia and osteoporosis are similarly lower using LMWH. The prolonged half-life of LMWH compared with its unfractionated counterpart permits less frequent dosing and less frequent thrombotic monitoring. Both promote patient acceptance and compliance.

Whereas LMWH has been shown to be safe, effective, and convenient in the treatment of many clotting disorders unrelated to reproduction,172 their use in pregnancy is only now being evaluated. Specific use of LMWH (in combination with low-dose ASA) among patients with RPL and thrombotic disorders has been reported. To date, the safety and efficacy of LMWH use have been suggested by interventional trials conducted among RPL patients with APAS169 and among those with activated protein C resistance associated with factor V Leiden.173,174,175

ASPIRIN PROPHYLAXIS.

Patients seeking therapy for reproductive disorders, including those experiencing isolated and recurrent spontaneous pregnancy losses, tend to be well-informed. They are therefore quick to apply the well-publicized cardiovascular benefits and infrequent side effects of daily prophylactic low-dose ASA to their own medical condition. Many patients with histories of recurrent loss are now either self-prescribing this therapy or inquiring about its efficacy. To summarize the fairly limited amount of data addressing prophylactic ASA use among patients with RPL, there is no good evidence for its clinical application either among patients with unexplained RPL or in those with coagulopathies. Although ASA/heparin has been demonstrated to be effective in the prevention and treatment of adverse pregnancy outcomes among patients with APAS (see above), the sole use of low-dose ASA among patients with RPL and APAS appears ineffective.168,176 According to one recent, large, prospective interventional trial, the use of prophylactic low-dose ASA cannot be recommended for the treatment of patients with unexplained recurrent early pregnancy losses (earlier than 13 weeks' gestation).177 Its use among women with unexplained late pregnancy losses (later than 13 weeks'gestation) did, however, appear promising.177

Immunomodulating Therapies

Other than the antithrombotic approaches, all alternative therapies for immune-mediated spontaneous pregnancy loss can be classified as immunoregulatory. Historical controversy over whether an immune response to the implanting pregnancy was essential or detrimental has led to the development and use of both immunosuppressive and immunostimulating therapies for the treatment of these patients. Although a large number of immune interventions have been proposed, these therapies remain experimental, and many have been proven ineffective; some even appear to be unsafe.

Although immune interactions seem to be involved in some instances of isolated spontaneous pregnancy loss, most of the published interventional studies involving immunotherapy for the prevention of subsequent pregnancy loss have been limited to patients with recurrent losses. Even these studies, however, have been limited by poor study design, small sample size, lack of patient prestratification by maternal age and number of prior losses before randomization, and a variety of methodologic and statistical inaccuracies. Another glaring omission in most studies has been the consistent analysis of aborted tissues for chromosomal abnormalities, because approximately 60% of individual losses are undoubtedly due to numeric chromosomal aberrations.178 Immunotherapy for RPL has been recently addressed in a number of excellent reviews179,180,181 and will be briefly summarized here.

Immunosuppressive Therapies

The hypothesis that inappropriate cellular or humoral immune responses to the implanting fetus cause pregnancy loss fostered the development of several immunosuppressive therapeutic approaches to the treatment of patients with immune-mediated RPL. These suppressive therapies have included the use of standard immunosuppressive drugs, immunosuppressive reproductive hormones, and intravenous immunoglobulins.

STANDARD IMMUNOSUPPRESSIVE THERAPIES.

Several immunoregulatory regimens, including the use of cyclosporine, pentoxifylline, and nifedipine, have been suggested based on related efficacy trials in the autoimmunity and transplantation literature. Extension of most of these interventions to patients with RPL, however, has been hampered by known or unknown maternal and fetal risks. For instance, plasmapheresis has been used for the prevention of thrombotic complications associated with the presence of antiphospholipid antibodies, and one isolated study has examined its efficacy among patients with recurrent abortion and antiphospholipid antibodies.182 The best-studied of the standard immunosuppressive approaches has involved the use of corticosteroid therapy. Although there exists evidence that prednisone is useful in the treatment of patients with recurrent pregnancy loss and APAS,183 antithrombotic therapy with heparin and aspirin has been shown to be effective and safer in this same patient population. A recent, large, randomized, placebo-controlled trial addressing the use of prednisone in combination with low-dose ASA among women with autoantibodies and RPL may relegate this therapy to historical interest only. In this investigation, pregnancy outcomes for treated and control patients were similar, but the incidence of maternal diabetes and hypertension and the risk of premature delivery were all increased among those treated with prednisone and ASA.184

IMMUNOSUPPRESSIVE REPRODUCTIVE HORMONES.

Both estrogen and progesterone have immunomodulating effects.82,83,84,85,86,87,88,89,90,91,92,93 The well-described and marked elevations of these reproductive hormones during pregnancy make them attractive candidates for use as immunosuppressive agents among patients with presumed immune-mediated pregnancy losses. Still, only progesterone has been tested for reproductive effects to date, and these investigations have limited clinical therapeutic applicability.185,186 As mentioned previously, in vitro evidence suggests that progesterone alters Th1/Th2-type cytokine responses,76,84,85,185,186 which might promote its appropriate use in patients with RPL and cytokine dysregulation at the maternal—fetal interface. Documenting such local dysregulation, however, is problematic. Further, even if it were possible to document, it is unclear whether systemic or local therapy would best address this dysregulation. Vaginal administration of progesterone may increase local, intrauterine concentrations of the hormone better than systemic administration and may therefore provide improved local immunosuppression while averting systemic side effects. A prospective, multicenter, placebo-controlled trial using progesterone in patients with RPL is underway and should provide much-needed insight into this intriguing therapeutic approach.

INTRAVENOUS IMMUNOGLOBULIN.

The pooling of the immunoglobulin proteins isolated from the serum of a large number of volunteer blood donors results in a blood product known as intravenous immunoglobulin (IVIg). The use of IVIg may be accurately categorized as immunosuppressive therapy, although the mechanistic basis for IVIg immunoregulation is poorly understood. Still, the list of potential immunoregulatory mechanisms is long and includes decreased autoantibody production and increased autoantibody clearance, T-cell and Fc receptor regulation,187 complement inactivation,188 enhanced T-cell suppressor function, decreased T-cell adhesion to the extracellular matrix,189 and downregulation of Th1 cytokine synthesis.190 Because IVIg is a blood product, its use is associated with a variety of real and potential risks. Like other blood products, IVIg is screened for transmissible agents such as HIV and hepatitis. However, up to 150 different donors are required to manufacture one vial of IVIg, and the transmission of prion-related disorders (e.g., “mad cow disease,” Jakob-Creutzfeldt disease) and other disorders that are not readily detected in routine screening should be considered a potential risk of therapy. Side effects of IVIg therapy include nausea, headache, myalgias, hypotension, and in some instances anaphylaxis.191

Therapy with IVIg for recurrent pregnancy loss is expensive, invasive, and time-consuming, requiring multiple intravenous infusions over the course of pregnancy. Although the list of financial and medical costs appears daunting, does treatment outcome using IVIg among patients with RPL justify the expense, side effects, and risks of this immunointervention? The data addressing the use of IVIg in patients with RPL is fairly extensive and, until recently, the answer to this question remained unclear. Although a number of relatively small studies using a variety of treatment protocols produced conflicting results, larger and more consistent studies have been unable to conclusively justify the use of IVIg in patients with unexplained (and presumed immunologic) RPL.192,193,194,195,196,197,198,199 Still, some may argue that the use of IVIg may be indicated among those specific patients with autoimmune-mediated pregnancy loss associated with APAS.200,201 Although they would be useful, no safety and efficacy studies have been performed comparing the use of IVIg with that of unfractionated heparin/ASA or LMWH/ASA in these patients.

Immunostimulating Therapies

The development of immunostimulating therapies for the prevention of spontaneous pregnancy losses was based on the premise that an appropriate immune recognition and reaction to the implanting fetus was essential to subsequent pregnancy maintenance. There are problems with this underling premise. First, although we know that pregnancies are specifically recognized and immune reactions mounted by the pregnant woman,123 a requirement for these responses has never been definitively demonstrated. Further, the fetal/paternal alloantigens recognized by the maternal immune system have never been appropriately isolated and identified. This explains the variety of antigen preparations and routes of administration that have been promoted during the development of immunostimulating therapy for RPL. For instance, syncytiotrophoblast microvillus plasma membrane vesicles have been prepared and administered intravenously to mimic the fetal cell contact with maternal blood that normally occurs in pregnancy.202 The erroneous belief that TLX was part of an idiotype/anti-idiotype control system203 led to the use of third-party seminal plasma suppositories for immunostimulation in women with histories thought to be consistent with immune-mediated RPL.204 Neither therapy was proven effective.

Leukocyte Immunization

The best-studied immunostimulation therapy to be promoted for use among patients with immune RPL involved the intravenous administration of either paternal or pooled donor leukocytes. Because leukocytes from either source supply innumerable alloantigens, it is not surprising that maternal immune responses would be stimulated. Immune mechanisms responsible for the transfer of these systemic responses to potential fetoprotective effects were suggested205,206,207,208 but never proven. Like IVIg, paternal or donor leukocytes represent blood products, and their administration to a third party carries the same risks as with other blood products. Reported risks specific to the use of leukocyte immunization in pregnant women encompass a variety of adverse fetal and maternal effects, including graft-versus-host disease, severe intrauterine growth restriction, and potentially fatal fetal thrombocytopenia.6,209,210,211,212,213,214 Again, as with the use of IVIg in RPL patients, the question arises of whether the treatment outcome with the use of leukocyte immunization among patients with RPL justifies the expense, side effects, and risks of this immunointervention. The data addressing this question are similarly heterogeneous and similarly confusing.209,210,215,216,217,218 However, the most recent and largest trial evaluating the efficacy of leukocyte immunization in patients with unexplained RPL was a part of the Recurrent Miscarriage Study (REMIS).218 This investigation involved more than 90 patients per treatment arm and was prospective, placebo-controlled, randomized, and double-blinded. The study indicated that paternal leukocyte immunization was not efficacious in couples with unexplained RPL; indeed, women receiving leukocyte immunization were more likely to experience a repeat loss than women receiving the placebo.

Back to Top
CONCLUSIONS

Immunologic interactions are complex. To those studying the immune interactions surrounding implantation, it is sobering to note that pregnancy does not appear to require an intact maternal immune system. To this point, women and animals who lack immunoglobulins (agammaglobulinemic) successfully reproduce.219 Women with severe immune deficiencies, mice that lack T and B cells (SCID mice), and mice with congenital absence of their thymus (nude mice) also carry pregnancies to term deliveries.220 Defining the role of a particular immune factor in pregnancy maintenance is therefore likely to be neither simple nor expedient.

The prognosis for a patient with RPL is good. Any given patient with a history of RPL is more likely to carry her next pregnancy to term than to miscarry. In fact, for patients with a history of RPL, the risk of subsequent pregnancy loss is estimated to be 24% after two clinically recognized losses, 30% after three losses, and 40% to 50% after four losses.221 These prognostic data, while helpful, still frustrate attempts to document the efficacy of interventional therapy. Unless interventions are remarkably effective, treatment groups must be almost prohibitively large to demonstrate alterations in outcome.

Clinical studies on immune-mediated pregnancy loss have been regrettably difficult to interpret. The disease itself is hard to accurately diagnose, typically relying solely on the exclusion of other etiologies. RPL represents a number of different specific disorders. With inconsistent clinical definition, trials involving these patients are difficult to compare and evaluate. Trial design is frequently substandard, with lack of rationale, lack of appropriate controls, and poor statistical analysis limiting the ability to draw rational conclusions from reported results.

The future, however, is not bleak. The rapid expansion in our understanding of the molecular and cellular immune environment at the maternal—fetal interface will surely stimulate clinical applications, and clinical insight will translate into therapeutic advances. The next decade will undoubtedly witness significant inroads into the diagnosis and treatment of immune-mediated isolated and recurrent spontaneous pregnancy loss.

Back to Top
REFERENCES

1. Edmonds DK, Lindsay KI, Miller JF: Early embryonic mortality in women. Fertil Steril 38: 447– 453, 1982

2. Alberman E: The epidemiology of repeated abortion. In Beard RW, Sharp F (eds): Early Pregnancy Loss: Mechanisms and Treatment, pp 9–17. New York, Springer-Verlag, 1988

3. Wilcox AJ, Weinberg CR, O'Connor JF, et al: Incidence of early loss of pregnancy. N Engl J Med 319: 189– 194, 1988

4. Hill JA: Recurrent pregnancy loss. In Creasy RK, Resnik R (eds): Maternal Fetal Medicine, pp 423–443. 3d ed. New York, WB Saunders, 1998

5. Stephenson MD: Frequency of factors associated with habitual abortion in 197 couples. Fertil Steril 66: 24– 29, 1996

6. Hill JA: Sporadic and recurrent spontaneous abortion. Curr Probl Obstet Gynecol Fertil 17: 114– 162, 1994

7. Summers PR: Microbiology relevant to recurrent miscarriage. Clin Obstet Gynecol 37: 722– 29, 1994

8. Paukku M, Tulppala M, Puolakkainen M, et al: Lack of association between serum antibodies to Chlamydia trachomatis and recurrent pregnancy loss. Fertil Steril 72: 427– 430, 1999

9. Robb J, Benirshke K, Barmeyer R: Intrauterine latent herpes simplex virus infection: spontaneous abortion. Hum Pathol 17: 1196– 1209, 1986

10. Altshuler G: Immunologic competence of the immature human fetus:Morphologic evidence from intra-uterine cytomegalovirus infection. Obstet Gynecol 43: 811– 816, 1974

11. Heyborne KD, Wilkin SS, McGregor JA: Tumor necrosis factor- α in midtrimester amniotic fluid is associated with impaired intrauterine fetal growth. Am J Obstet Gynecol 167: 920– 925, 1992

12. Romero R, Mazor M, Sepulueda W et al: Tumor necrosis factor in preterm and term labor. Am J Obstet Gynecol 166: 1576– 1587, 1992

13. Furman MH, Ploegh HL, Schust DJ: Can viruses help us to understand and classify the MHC class I molecules at the maternal-fetal interface? Hum Immunol 61: 1169– 1176, 2000

14. Thellin O, Coumans B, Zorzi W et al: Tolerance of the feto-placental “graft”: Ten ways to support a child for nine months. Curr Opin Immunol 12: 731– 737, 2000

15. Abbas AK, Lichtman AH, Pober JS: Cellular and Molecular Immunology. 4th ed. Philadelphia, WB Saunders, 2000

16. Lanier LL: Activating and inhibitory NK cell receptors. Ann Rev Immunol 452: 13– 18, 1998

17. Lefrancois L, Fuller B, Huleatt JW et al: On the front lines:intraepithelial lymphocytes as primary effectors of intestinal immunity. Springer Semin Immunopathol 18: 463– 475, 1997

18. Gould DS, Ploegh HL, Schust DJ: Murine female reproductive tract intraepithelial lymphocytes display selection characteristics distinct from both peripheral and other mucosal T cells. J Reprod Immunol 52: 85, 2001

19. Loke YW, King A: Immunological aspects of human implantation. J Reprod Fertil Steril 55: 83– 90, 2000

20. Johnson PM, Christmas PE, Vince GS: Immunological aspects of implantation and implantation failure. Hum Reprod 14 (suppl 2): 26– 36, 1999

21. Vince GS, Johnson PM: Leukocyte populations and cytokine regulation in human uteroplacental tissues. Biochem Soc Trans 28: 191– 195, 2000

22. Loke YW, King A: Decidual natural-killer-cell interaction with trophoblast: Cytolysis or cytokine production? Biochem Soc Trans 28: 196– 198, 2000

23. King A, Hiby SE, Gardner L et al: Recognition of trophoblast HLA class I molecules by decidual NK cell receptors—a review. Placenta 21 (suppl A): S81– 85, 2000

24. Godfrey DI, Hammond KJ, Poulton LD et al: NKT cells: facts, functions, and fallacies. Immunol Today 21: 573– 83, 2000

25. Ito K, Karasawa M, Kawano T et al: Involvement of decidual Valpha14 NKT cells in abortion. Proc Natl Acad Sci USA 97: 740– 744, 2000

26. Dang Y, Heyborne KD: Cutting edge: Regulation of uterine NKT cells by a fetal class I molecule other than CD1. J Immunol 166: 3641– 3644, 2001

27. Dang Y, Beckers J, Wang CR, Heyborne KD: Natural killer 1. 1(+) alpha beta T cells in the periimplantation uterus. Immunology 101: 484– 491, 2000

28. Mincheva-Nilsson L, Baranov V, Yeung M et al: Immunomorphologic studies of human decidua-associated lymphoid cells in normal early pregnancy. J Immunol 152: 2020– 2032, 1994

29. Christmas SE, Brew R, Thornton SM et al: Extensive TCR junctional diversity of V gamma 9/V delta 2 clones from human reproductive tract tissues. J Immunol 155: 2453– 2458, 1995

30. Vassiliadou N, Bulmer JN: Characterization of endometrial T lymphocyte subpopulations in spontaneous early pregnancy loss. Hum Reprod 13: 44– 47, 1998

31. Hayday AC: γ δ cells: A right time and a right place for a conserved third way of protection. Ann Rev Immunol 18: 975– 1026, 2000

32. Chang MD, Pollard JW, Khalili H et al: Mouse placental macrophages have a decreased ability to present antigen. Proc Natl Acad Sci USA 90: 462– 466, 1993

33. Clifford K, Flanagan AM, Regan L: Endometrial CD56+ natural killer cellls in women with recurrent miscarriage: A histomorphometric study. Hum Reprod 14: 2727– 2730, 1999

34. Quenby S, Bates M, Doig T et al: Pre-implantation endometrial leukocytes in women with recurrent miscarriage. Hum Reprod 14: 2836– 2891, 1999

35. Yamamoto T, Takahashi Y, Kase N, Mori H: Proportion of CD56+3+ T cells in decidual and peripheral lymphocytes of normal pregnancy and spontaneous abortion with and without history of recurrent abortion. Am J Reprod Immunol 42: 355– 360, 1999

36. Yamamoto T, Takahashi Y, Kase N, Mori H: Decidual natural killer cells in recurrent spontaneous abortion with normal chromosomal content. Am J Reprod Immunol 41: 337– 342, 1999

37. Lachapelle MH, Miron P, Hemmings R, Roy DC: Endometrial T, B, and NK cells in patients with recurrent spontaneous abortion. Altered profile and pregnancy outcome. J Immunol 156: 4027– 4034, 1996

38. Pudney J, Quayle AJ, Anderson DJ: Personal communication, 2001

39. Schon MP, Arya A, Murphy EA et al: Mucosal T lymphocyte numbers are selectively reduced in integrin αE (CD103)-deficient mice. J Immunol 162: 6641– 6649, 1999

40. Kruse A, Hallman R, Butcher EC: Specialized patterns of vascular differentiation antigens in the pregnant mouse uterus and the placenta. Biol Reprod 61: 1393– 1401, 1999

41. Perry LL, Feilzer K, Portis JL, Caldwell HD: Distinct homing pathways direct T lymphocytes to the genital and intestinal mucosae in Chlamydia -infected mice. J Immunol 160: 2905– 2914, 1998

42. Pudney J, Anderson DJ: Immunobiology of the human penile urethra. Am J Pathol 147: 155– 165, 1995

43. Billington WD, Davies M, Bell SC: Maternal antibody to foetal histocompatibility and trophoblast-specific antigens. Ann Immunol (Paris) 135D: 331, 1984

44. Tafuri A, Alferink J, Moller P et al: T cell awareness of paternal alloantigens during pregnancy. Science 270: 630– 633, 1995

45. Sargent IL, Wilkins T, Redman CWG: Maternal immune responses to the fetus in early pregnancy and recurrent miscarriage. Lancet 2: 1099– 1104, 1994

46. Bermas BL, Hill JA: Proliferative responses to recall antigens are associated with pregnancy outcome in women with a history of recurrent spontaneous abortion. J Clin Invest 100: 1330– 1334, 1997

47. Stewart CL, Kaspar P, Brunet LJ et al: Blastocyst implantation depends on maternal expression of leukemia inhibiting factor. Nature 358: 76– 79, 1992

48. Chen JR, Cheng JG, Shatzer T et al: Leukemia inhibiting factor can substitute for nidatory estrogen and is essential to inducing a receptive uterus for implantation but is not essential for subsequent embryogenesis. Endocrinology 141: 4365– 4372, 2000

49. Asnagli H, Murphy KM: Stability and commitment in T helper cell development. Curr Opin Immunol 13: 242– 247, 2001

50. Palmer EM, Seventer GA: Human helper T cell differentiation is regulated by the combined action of cytokines and accessory cell-dependent co-stimulatory signals. J Immunol 158: 2654– 2662, 1997

51. O'Garra A, Arai N: The molecular basis of T helper 1 and T helper 2 cell differentiation. Trends Cell Biol 10: 542– 550, 2000

52. Kurt-Jones EA, Hamberg S, Ohara J et al: Heterogeneity of helper/inducer T lymphocytes. I. Lymphokine production and lymphokine responsiveness. J Exp Med 166: 1774– 1787, 1987

53. Romagnani S: Human TH1 and TH2 subsets: Doubt no more. Immunol Today 8: 256– 257, 1991

54. Mosmann TR, Coffman RL: Heterogeneity of cytokine secretion patterns and functions of helper T cells. Adv Immunol 46: 111– 147, 1989

55. Ramagnani S: Human TH1 and TH2 subsets: Regulation of differentiation and role in protection and immunopathology. Int Arch Allergy Immunol 4: 279– 285, 1992

56. Mosmann TR, Coffman RL: TH1 and TH2 cells: Different patterns of lymphokine secretion lead to different functional properties. Annu Rev Immunol 7: 145– 173, 1989

57. Maggi E, Parronchi P, Monetti R et al: Reciprocal regulatory effects of IFN- α and IL-4 in in vitro development of human TH1 and TH2 clones. J Immunol 148: 2142– 2147, 1992

58. Fiorentino DF, Bond MW, Mosmann TR: Two types of mouse T helper cell IV. TH2 clones secrete a factor that inhibits cytokine production by TH1 clones. J Exp Med 170: 2081– 2095, 1989

59. Mosmann TR, Moore KW: The role of IL-10 in cross regulation of TH1 and TH2 responses. Immunol Today 12: A49– 53, 1991

60. Sen DR, Fox H: The lymphoid tissue of the endometrium. Gynecol Pathol 163: 371, 1967

61. Kearns M, Lala PH: Bone marrow origin of decidual cell precursors in the pseudopregnant mouse uterus. J Exp Med 155: 1537– 1554, 1982

62. Bulmer JN, Sunderland CA: Immunohistological characterization of lymphoid cell populations in the early human placental bed. Immunology 52: 349– 357, 1984

63. Tabibzadeh S: Human endometrium: An active site of cytokine production and action. Endocr Rev 12: 272– 290, 1991

64. Klentzeris LD, Bulmer JN, Warren A et al: Endometrial lymphoid tissue in the timed endometrial biopsy: Morphometric and immunohistochemical aspects. Am J Obstet Gynecol 167: 667– 674, 1992

65. Hill JA, Polgar K, Anderson DJ: T helper 1-type immunity to trophoblast antigens in women with recurrent spontaneous abortion. JAMA 273: 1933– 1936, 1995

66. Hill JA, Polgar K, Harlow BL, Anderson DJ: Evidence of embryo- and trophoblast-toxic cellular immune response(s) in women with recurrent spontaneous abortion. Am J Obstet Gynecol 166: 1044– 1052, 1992

67. Mallmann P, Werner A, Krebs D: Serum levels of interleukin-2 and tumor necrosis factor- α in women with recurrent abortion. Am J Obstet Gynecol 163: 1367, 1990

68. Yamada H, Polgar K, Hill JA: Cell-mediated immunity to trophoblast antigens in women with recurrent spontaneous abortion. Am J Obstet Gynecol 170: 1339– 1344, 1994

69. Berkowitz RS, Hill JA, Kurtz CB, Anderson DJ: Effects of products of activated leukocytes (lymphokines and monokines) on the growth of malignant trophoblast cells in vitro. Am J Obstet Gynecol 158: 199– 204, 1988

70. Hunt JS, Soales MJ, Lei MG et al: Products of lipopolysaccharide-activated macrophages (tumor necrosis factor- α, transforming growth factor- β) but not lipopoly-saccharide modify DNA synthesis by rat trophoblast cells exhibiting the 80-KDa LPS-binding protein. J Immunol 143: 1606– 1613, 1989

71. Hill J, Haimovici F, Anderson DJ: Products of activated lymphocytes and macrophages inhibit mouse embryo development in vitro. J Immunol 139: 2250– 2254, 1987

72. Ecker JL, Laufer MR, Hill JA: Measurement of embryotoxic factors is predictive of pregnancy outcome in women with a history of recurrent abortion. Obstet Gynecol 81: 84– 87, 1993

73. Lea RG, Tulppala M, Critchley HO: Deficient syncytiotrophoblast tumor necrosis factor-alpha characterizes failing first trimester pregnancies in a subgroup of recurrent miscarriage patients. Hum Reprod 12: 1313– 1320, 1997

74. von Wolff M, Thaler CJ, Strowitzki T et al: Regulated expression of cytokines in the human endometrium throughout the menstrual cycle:Dysregulation in habitual abortion. Mol Hum Reprod 6: 627– 634, 2000

75. Lim KJ, Odukoya OA, Ajjan RA et al: The role of T-helper cytokines in human reproduction. Fertil Steril 73: 136– 142, 2000

76. Piccini MP, Beloni L, Livi C et al: Defective production of both leukemia inhibiting factor and type 2 cytokine production by decidual T cells in unexplained recurrent abortion. Nat Med 4: 1020– 1024, 1998

77. Raghupathy R, Makhseed M, Azizieh F et al: Cytokine production by maternal lymphocytes during normal human pregnancy and in unexplained recurrent spontaneous abortion. Hum Reprod 15: 713– 718, 2000

78. Raghupathy R, Makhseed M, Azizieh F et al: Maternal Th1- and Th2-type reactivity to placental antigens in normal human pregnancy and unexplained recurrent spontaneous abortions. Cell Immunol 196: 122– 130, 1999

79. Schust DJ, Hill JA: Correlation of serum cytokine and adhesion molecule determinations with pregnancy outcome. J Soc Gynecol Invest 3: 259– 261, 1996

80. Makhseed M, Raghupathy R, Azizieh F et al: Circulating cytokines and CD30 in normal human pregnancies and recurrent spontaneous abortions. Hum Reprod 15: 2011– 2017, 2000

81. Jenkins C, Roberts J, Wilson R et al: Evidence of a T(H)1 type response associated with recurrent miscarriage. Fertil Steril 73: 1206– 1208, 2000

82. Grossman CJ: Possible underlying mechanisms of sexual dimorphism in the immune response: fact and hypothesis. J Steroid Biochem 34: 241– 251, 1989

83. Siiteri PK, Febres F, Clemens LE et al: Progesterone and maintenance of pregnancy. Is progesterone nature's immunosuppressant? Ann NY Acad Sci 286: 384– 397, 1977

84. Vassiliadou N, Tucker L, Anderson DJ: Progesterone-induced inhibition of chemokine receptor expression on peripheral blood mononuclear cells correlates with reduced HIV-1 infectability in vitro. J Immunol 14: 2252– 2256, 1999

85. Hunt JS, Miller L, Roby KF et al: Female steroid hormones regulate production of pro-inflammatory molecules in uterine leukocytes. J Reprod Immunol 35: 87– 99, 1997

86. Knoferi MW, Diodato MD, Angele MK et al: Do female sex steroids adversely or beneficially effect the depressed immune responses in males after trauma-hemorrhage? Arch Surg 135: 425– 433, 2000

87. Gregory MS, Duffner LA, Faunce DE, Kovacs EJ: Estrogen mediates the sex difference in post-burn immunosuppression. J Endocrinol 164: 129– 138, 2000

88. Muller V, Szabo A, Viklicky O et al: Sex hormones and gender-related differences: their influence on chronic renal allograft rejection. Kidney Int 55: 2011– 2020, 1999

89. Salem ML, Matsuzaki G, Kishihara K et al: Beta-estradiol suppresses T cell-mediated delayed-type hypersensitivity through suppression of antigen-presenting cell function and Th1 induction. Int Arch Allergy Immunol 121: 161– 169, 2000

90. Correale J, Arias M, Gilmore W: Steroid hormone regulation of cytokine secretion by proteolipid protein-specific CD4+ T cell clones isolated from multiple sclerosis patients and normal control subjects. J Immunol 161: 3365– 3374, 1998

91. Cutolo M, Wilder RL: Different roles for androgens and estrogens in the susceptability to autoimmune rheumatic diseases. Rheum Dis Clin North Am 26: 825– 839, 2000

92. Kanik KS, Wilder RL: Hormonal alterations in rheumatoid arthritis. Rheum Dis Clin North Am 26: 825– 839, 2000

93. Zaffaroni M, Ghezzi A: The prognostic value of age gender, pregnancy and endocrine factors in multiple sclerosis. Neurol Sci 21 (suppl 2): S857– 860, 2000

94. Chapman SJ: Varicella in pregnancy. Semin Perinatol 22: 339– 346, 1998

95. Runnebaum B, Stober I, Zander J: Progesterone, 20 α -dihydroprogesterone, and 20 β -dihydroprogesterone in mother and child at birth. Acta Endocrinol 80: 569– 576, 1975

96. Munn DH, Shafizadeh E, Attwood JT et al: Inhibition of T cell proliferation by macrophage tryptophan metabolism. J Exp Med 189: 1363– 1372, 1999

97. Mellor AL, Munn DH: Immunology at the maternal-fetal interface: lessons from T cell tolerance and suppression. Ann Rev Immunol 18: 367– 391, 2000

98. Munn DH, Zhou M, Atwood JT et al: Prevention of allogeneic fetal rejection by tryptophan catabolism. Science 281: 1191– 1193, 1998

99. Meier AH, Wilson JM: Tryptophan feeding adversely influences pregnancy. Life Sci 32: 1193– 1196, 1983

100. Kamimura S, Eguchi K, Yonezawa M, Sekiba K: Localization and developmental change of indoleamine 2,3-dioxygenase activity in the human placenta. Acta Med Okayama 45: 135– 139, 1991

101. Schrocksnadel H, Baier-Bitterlich G, Dapunt O et al: Decreased plasma tryptophan in pregnancy. Obstet Gynecol 88: 47– 50, 1996

102. Mattson R: The non-expression of MHC class II in trophoblast cells. Am J Reprod Immunol 40: 385– 394, 1998

103. Murphy SP, Tomasi TB: Absence of MHC class II antigen expression in trophoblast cells results from a lack of class II transactivator (CIITA) gene expression. Mol Reprod Dev 51: 1– 12, 1998

104. Kovats S, Main EK, Librach C et al: A class I antigen HLA-G expressed in human trophoblasts. Science 248: 220– 223, 1990

105. Ellis SA, Palmer MS, McMichael AJ: Human trophoblast and the choriocarcinoma cell line BeWo express a truncated HLA class I molecule. J Immunol 144: 731– 735, 1990

106. Wei X, Orr HT: Differential expression of HLA-E, -F, and -G transcripts in human tissue. Hum Immunol 29: 131– 142, 1990

107. Sernee MF, Ploegh HL, Schust DJ: Why certain antibodies cross-react with HLA-A and HLA-G: epitope mapping of two common MHC class I reagents. Mol Immunol 35: 177– 188, 1998

108. King A, Burrows TD, Hiby SE et al: Surface expression of HLA-C antigen by human extravillous cytotrophoblast. Placenta 21: 376– 387, 2000

109. King A, Allen DS, Bowen M et al: HLA-E is expressed on trophoblast and interacts with CD94/NKG2 receptors on decidual NK cells. Eur J Immunol 30: 1623– 1631, 2000

110. Zhou Y, Fisher SJ, Janatpour M et al: Human cytotrophoblasts adopt a vascular phenotype as they differentiate. J Clin Invest 99: 2139– 2151, 1997

111. Kam EP, Gardner L, Loke YW, King A: The role of trophoblast in the physiological changes in the decidual spiral arteries. Hum Reprod 14: 2131– 2138, 1999

112. Damsky CH, Fisher SJ: Trophoblast pseudo-vasculogenesis: Faking it with endothelial adhesion receptors. Curr Opin Cell Biol 10:660–666, 1998.

113. Hamai Y, Fujii T, Yamashita T et al: Peripheral blood mononuclear cells from women with recurrent spontaneous abortion exhibit an aberrant reaction to release cytokines upon the direct contact of human leukocyte antigen-G-expressing cells. Am J Reprod Immunol 40: 408– 413, 1998

114. Kanai T, Fujii T, Unno N et al: Human leukocyte antigen-G-expressing cells differently modulate the release of cytokines from mononuclear cells present in the decidua versus peripheral blood. Am J Reprod Immunol Microbiol 45: 94– 99, 2001

115. Lim KH, Zhou Y, Janatpour M et al: Human cytotrophoblast differentiation/invasion is abnormal in pre-eclampsia. J Clin Invest 99: 2139– 2151, 1997

116. Goldman-Wohl DS, Ariel I, Greenfield C et al: Lack of human leukocyte antigen-G expression in extravillous trophoblasts is associated with pre-eclampsia. Mol Hum Reprod 6: 88– 95, 2000

117. Hunt JS, Jadhav L, Chu W et al: Soluble HLA-G circulates in maternal blood during pregnancy. Am J Obstet Gynecol 183: 682– 688, 2000

118. Yamashita T, Fujii T, Tokunaga K et al: Analysis of human leukocyte antigen-G polymorphism including intron 4 in Japanese couples with habitual abortion. Am J Reprod Immunol 41: 159– 163, 1999

119. Steffensen R, Christiansen OB, Bennett EP, Jerslid C: HLA-E polymorphism in patients with recurrent spontaneous abortion. Tissue Antigens 52: 569– 572, 1998

120. Feinman MA, Kliman JH, Main EK: HLA antigen expression and induction by a-interferon in cultured human trophoblast. Am J Obstet Gynecol 157: 1429– 1434, 1987

121. Hill JA: Immunological mechanisms of pregnancy maintenance and failure:a critique of theories and therapy. Am J Reprod Immunol 22: 33– 42, 1990

122. Hill JA, Melling GC, Johnson PM: Immunohistochemical studies of human uteroplacental tissues from first trimester spontaneous abortion. Am J Obstet Gynecol 173: 90– 96, 1995

123. Eblen AC, Gercel-Taylor C, Shields LB et al: Alterations in humoral immune responses associated with recurrent pregnancy loss. Fertil Steril 73: 305– 313, 2000

124. Rocklin RE, Kitzmiller JL, Garvey MR: Maternal-fetal relation: Further characterization of an immunologic blocking factor that develops during pregnancy. Clin Immunol Immunopathol 22: 305– 315, 1982

125. Amos DB, Kostyn DD: HLA: A central immunological agency of man. Adv Hum Genet 10: 137– 141, 1980

126. Coulam CB: Immunological tests in the evaluation of reproductive disorders: A critical review. Am J Obstet Gynecol 167: 1844– 1851, 1992

127. McIntyre JA, Faulk WP, Verhulst ST, Colliver JA: Human trophoblast-lymphocyte cross-reactive (TLX) antigens define a new alloantigen system. Science 222: 1135– 1137, 1983

128. Roumen G, Roussev MD, Vanderpuye OA et al: A role for TLX antigens in pregnancy. Acta Eur Fertil 22: 181– 187, 1991

129. Bjercke S: Recurrent abortion and lymphocyte transfusions. Acta Obstet Gynecol Scand 73: 373– 376, 1994

130. Purcell DF, McKenzie IF, Lublin DM et al: The human cell surface glycoproteins Hu Ly-M5, membrane co-factor protein (MCP) of the complement system, and trophoblast leukocyte common (TLX) antigen are CD46. Immunology 70: 155– 161, 1990

131. Beer AE, Quebbeman JF, Ayers JW, Haines RF: Major histocompatibility complex antigens, maternal and paternal immune responses and chronic habitual abortion in humans. Am J Obstet Gynecol 141: 987– 999, 1981

132. McIntyre JA, Faulk WP: Recurrent spontaneous abortion in human pregnancy: Results of immunogenetical, cellular and humoral tests. Am J Reprod Immunol 4: 165– 170, 1983

133. Ober CL, Martin AO, Simpson JL et al: Shared HLA antigens and reproductive performance among Hutterites. Am J Hum Genet 35: 994– 1004, 1983

134. Ober C, Hyslop T, Elias S et al: Human leukocyte antigen matching and fetal loss: Results of a 10-year prospective study. Hum Reprod 13: 33– 38, 1998

135. Christiansen OB, Anderson HH, Hojbjerre M et al: Maternal HLA class II allogenotypes are markers for the predisposition to fetal losses in families of women with unexplained recurrent fetal loss. Eur J Immunogenet 22: 323– 334, 1995

136. Christiansen OB, Rasmussen KL, Jerslid C, Grunnet N: HLA class II alleles confer susceptibility to recurrent fetal losses in Danish women. Tissue Antigens 44: 225– 233, 1994

137. Kutteh WH, Yetman DL, Carr AC et al: Increased prevalence of antithyroid antibodies identified in women with recurrent pregnancy loss but not in women undergoing assisted reproduction. Fertil Steril 71: 843– 848, 1999

138. Bussen SS, Steck T: Thyroid antibodies and their relation to antithrombin antibodies, anticardiolipin antibodies and lupus anticoagulant in women with recurrent spontaneous abortion (antithyroid, anticardiolipin and antithrombin autoantibodies and lupus anticoagulant in habitual aborters). Eur J Obstet Gynecol Reprod Biol 74: 139– 143, 1997

139. Esplin MS, Branch DW, Silver R, Stagnaro-Green A: Thyroid autoantibodies are not associated with recurrent pregnancy loss. Am J Obstet Gynecol 179: 1583– 1586, 1998

140. Rushworth FH, Backos M, Rai R et al: Prospective pregnancy outcome in untreated recurrent miscarriers with thyroid antibodies. Hum Reprod 15: 1637– 1639, 2000

141. Harris EN: Syndrome of the black swan. Br J Rheumatol 26: 324– 326, 1986

142. Wilson WA, Gharavi AE, Koike T et al: International consensus statement on preliminary classification criteria for definite antiphospholipid syndrome: report of an international workshop. Arthritis Rheum 42: 1309– 1311, 1999

143. Lockshin MD, Sammaritano LR, Schwartzman S: Validation of the Sapporo criteria for antiphospholipid syndrome. Arthritis Rheum 43: 440– 443, 2000

144. Out HJ, Bruinse HW, Christiaens, GC et al: A prospective, controlled multicenter study of the obstetric risks of pregnant women with antiphosphalipid antibodies. Br J Obstet Gynaecol 167: 26– 32, 1992

145. Kutteh WH, Lyda EC, Abraham SM, Wacholtz MC: Association of anticardiolipin antibodies and pregnancy loss in women with systemic lupus erythematosus Fertil Steril 60:449–455, 1993

146. Hanly JG, Gladman DD, Rose TH et al: Lupus pregnancy: a prospective study of placental changes. Arthritis Rheum 31: 358– 366, 1988

147. Lockshin MD, Druzin ML, Goei S et al: Antibody to cardiolipin as a predictor of fetal distress or death in pregnant patients with systemic lupus erythematosus. N Engl J Med 313: 152– 156, 1985

148. Harris EN, Asherson RA, Gharavi AE et al: Thrombocytopenia in SLE and related disorders: association with anticardiolipin antibodies. Br J Haematol 59: 227– 230, 1985

149. Cariou R, Tobelem G, Soria C, Caen J: Inhibition of protein C activation by endothelial cells in the presence of lupus anticoagulant. N Engl J Med 314: 1193– 1194, 1986

150. Freyssinet JM, Wiesel ML, Gauchy J et al: An IgM lupus anticoagulant that neutralizes the enhancing effect of phospholipid on purified endothelial thrombomodulin activity: A mechanism for thrombosis. Thromb Haemost 55: 309– 313, 1986

151. Rand JH, Wu X-X, Guller S et al: Reduction of annexin-V (placental anticoagulant protein-I) on placental villae of women with antiphospholipid antibodies and recurrent spontaneous abortion. Am J Obstet Gynecol 171: 1566– 1572, 1994

152. Rand JH, Wu X-X, Andree HA et al: Pregnancy loss in the antiphospholipid-antibody syndrome: A possible thrombogenic mechanism. N Engl J Med 337: 154– 160, 1997

153. Lyden TW, Ng AK, Rote NJ: Modulation of phosphatidyl-serine epitope expression on BeWo cells during forskolin treatment. Placenta 14: 177– 186, 1993

154. Bulla R, de Guarrini F, Pausa M et al: Inhibition of trophoblast adhesion to endothelial cells by the sera of women with recurrent spontaneous abortion. Am J Reprod Immunol 42: 116– 123, 1999

155. Galli M, Comfurius P, Maassen C et al: Anticardiolipin antibodies (ACA) directed not to cardiolipin but to plasma protein cofactor. Lancet 335: 1544– 1547, 1990

156. McNeil HP, Simpson RJ, Chesterman CN, Krillis SA: Anti-phospholipid antibodies are directed against a complex antigen that includes a lipid-binding inhibitor of coagulation: beta 2-glycoprotein I (apolipoprotein H). Proc Natl Acad Sci USA 87: 4120– 4124, 1990

157. Chamley LW, Allen JL, Johnson PM: Synthesis of beta2 glycoprotein 1 by the human placenta. Placenta 18: 403– 410, 1997

158. Lee RM, Emlen W, Scott JR et al: Anti-beta2-glycoprotein I antibodies in women with recurrent spontaneous abortion, unexplained fetal death, and antiphospholipid syndrome. Am J Obstet Gynecol 181: 642– 648, 1999

159. Ogasawara M, Aoki K, Katano K et al: Prevalence of autoantibodies in patients with recurrent miscarriage. Am J Reprod Immunol 41: 86– 90, 1999

160. Yetman DL, Kutteh WH: Antiphospholipid antibody panels and recurrent pregnancy loss: Prevalence of anticardiolipin antibodies compared with other antiphospholipid antibodies. Fertil Steril 66: 540– 546, 1996

161. Branch DW, Silver R, Pierangeli S et al: Antiphospholipid antibodies other than lupus anticoagulant and anticardiolipin antibodies in women with recurrent pregnancy loss, fertile controls, and antiphospholipid syndrome. Obstet Gynecol 89: 549– 555, 1997

162. Gris JC, Quere I, Sanmarco M et al: Antiphospholipid and antiprotein syndromes in non-thrombotic, non-autoimmune women with unexplained recurrent primary early foetal loss. The Nimes Obstetricians and Haematologists Study—NOHA. Thromb Haemost 84: 228– 236, 2000

163. Mavragani CP, Ionnidis JP, Tzioufas AG et al: Recurrent pregnancy loss and autoantibody profile in autoimmune diseases. Rheumatology (Oxford) 38:1228–1233, 1999

164. Hill JA, Choi BC: Maternal immunological aspects of pregnancy success and failure. J Reprod Fertil [Suppl] 55: 91– 97, 2000

165. Aoki K, Kajiura S, Matsumoto Y et al: Preconceptional natural-killer-cell activity as a predictor of miscarriage. Lancet 345: 1340– 1342, 1995

166. Emmer PM, Nelen WL, Steegers EA et al: Peripheral natural killer cell cytotoxicity and CD56(pos)CD16(pos) cells increase during early pregnancy in women with a history of recurrent spontaneous abortion. Hum Reprod 15: 1163– 1169, 2000

167. Ermel LD, Marshburn PB, Kutteh WH: Interaction of heparin with antiphospholipid antibodies (APA) from the sera of women with recurrent pregnancy loss. Am J Reprod Immunol 33: 14– 20, 1995

168. Kutteh WH: Antiphosphlipid antiboby-associated recurrent pregnancy loss: Treatment with heparin and low-dose aspirin is superior to aspirin alone. Am J Obstet Gynecol 174: 1584– 1589, 1996

169. Lima F, Khamashta MA, Buchanan NM et al: A study of sixty pregnancies in patients with the antiphospholipid syndrome. Clin Exp Rheumatol 14: 131– 136, 1996

170. Rai R, Cohen H, Dave M, Regan L: Randomised controlled trial of aspirin and aspirin plus heparin in pregnant women with recurrent miscarriage associated with phospholipid antibodies (or antiphospholipid antibodies). Br Med J 314: 253– 257, 1997

171. Bates SM, Ginsberg JS: Anticoagulation in pregnancy. Pharm Pract Manag Q 19: 51– 60, 1999

172. Bijsterveld NR, Hettiarachchi R, Peters R et al: Low-molecular-weight heparins in venous and arterial thrombotic disease. Thromb Haemost 82 (Suppl 1): 139– 147, 1999

173. Bar J, Cohen-Sacher B, Hod M et al: Low-molecular-weight heparin for thrombophilia in pregnant women. Int J Gynaecol Obstet 69: 209– 213, 2000

174. Younis JS, Ohel G, Brenner B et al: The effect of thromboprophylaxis on pregnancy outcome in patients with recurrent pregnancy loss associated with factor V Leiden mutation. Br J Obstet Gynaecol 107: 415– 419, 2000

175. Brenner B, Hoffman R, Blumenfeld Z et al: Gestational outcome in thrombophilic women with recurrent pregnancy loss treated with enoxaparin. Thromb Haemost 83: 693– 697, 2000

176. Pattison NS, Chamley LW, Birdsall M et al: Does aspirin have a role in improving pregnancy outcome for women with the antiphospholipid syndrome? A randomized controlled trial. Am J Obstet Gynecol 183: 1008– 1012, 2000

177. Rai R, Backos M, Baxter N et al: Recurrent miscarriage: An aspirin a day? Hum Reprod 15: 2220– 2223, 2000

178. Boue J, Boue A, Laser P: Retrospective and prospective epidemiologic studies of 1,500 karyotyped spontaneous abortions. Teratology 11: 11– 26, 1975

179. Hill JA: Immunotherapy for recurrent pregnancy loss: “Standard of care or buyer beware.” J Soc Gynecol Invest 4: 267– 273, 1997

180. Stovall DW, Van Voorhis BJ: Immunologic tests and treatments in patients with unexplained infertility, IVF-ET, and recurrent pregnancy loss. Clin Obstet Gynecol 42: 979– 1000, 1999

181. Kutteh WH: Recurrent pregnancy loss: An update. Curr Opin Obstet Gynecol 11: 435– 439, 1999

182. Ferro D, Quintarelli C, Russo G et al: Successful removal of antiphospholipid antibodies using repeated plasma exchanges and prednisone. Clin Exp Rheumatol 7: 103– 104, 1989

183. Lubbe WF, Butler WS, Palmer SJ, Liggins GC: Fetal survival after prednisone suppression of maternal lupus-anticoagulant. Lancet 1: 1361– 1363, 1983

184. Laskin CA, Bombardier C, Hannah ME et al: Prednisone and aspirin in women with autoantibodies and unexplained recurrent fetal loss. N Engl J Med 337: 148– 153, 1997

185. Polgar K, Hill JA: Progesterone inhibits in vitro embryotoxic factor production in women with recurrent spontaneous abortion. Proc Soc Gynecol Invest 41: 22– 26, 1994

186. Choi BC, Polgar K, Xiao L, Hill JA: Progesterone inhibits in vitro embryotoxic Th1 cytokine production to trophoblast in women with recurrent pregnancy loss. Hum Reprod 15 (Suppl 1): 46– 59, 2000

187. Samuelsson A, Towers TL, Ravetch JV: Anti-inflammatory activity of IVIG mediated through the inhibitory Fc receptor. Science 291: 484– 486, 2001

188. Mollnes TE, Hogasen K, De Carolis C et al: High-dose intravenous immunoglobulin treatment activates complement in vivo. Scand J Immunol 48: 312– 317, 1998

189. Jerzak M, Gorski A, Jerzak M et al: Intravenous immunoglobulin therapy influences T cell adhesion to extracellular matrix in women with a history of recurrent spontaneous abortion. Am J Reprod Immunol Microbiol 44: 336– 341, 2000

190. Dwyer JM: Manipulating the immune system with immunoglobulin. N Engl J Med 326: 107– 161, 1992

191. Thornton CA, Ballow M: Safety of intravenous immunoglobulin. Arch Neurol 50: 135– 136, 1993

192. Müeller-Eckhart G, Huni O, Poltrin B: IVIG to prevent recurrent spontaneous abortion. Lancet 1: 424– 425, 1991

193. Stricker RB, Steinleitner A, Bookoff CN et al: Successful treatment of immunologic abortion with low-dose intravenous immunoglobulin. Fertil Steril 73: 536– 540, 2000

194. Jablonowska B, Selbing A, Palfi M et al: Prevention of recurrent spontaneous abortion by intravenous immunoglobulin: a double-blind placebo-controlled study. Hum Reprod 14: 838– 841, 1999

195. Daya S, Gunby J, Porter F et al: Critical analysis of intravenous immunoglobulin therapy for recurrent miscarriage. Hum Reprod Update 5: 475– 482, 1999

196. Stephenson MD, Dreher K, Houlihan E, Wu V: Prevention of unexplained recurrent spontaneous abortion using intravenous immunoglobulin: A prospective, randomized, double-blinded, placebo-controlled trial. Am J Reprod Immunol 39: 82– 88, 1998

197. Christiansen OB: Intravenous immunoglobulin in the prevention of recurrent spontaneous abortion: The European experience. Am J Reprod Immunol 39: 77– 81, 1998

198. Perino A, Vassiliadis A, Vucetich A et al: Short-term therapy for recurrent abortion using intravenous immunoglobulins: Results of a double-blind placebo-controlled Italian study. Hum Reprod 12: 2388– 2392, 1997

199. Coulam CB, Krysa L, Stern JJ, Bustillo M: Intravenous immunoglobulin for treatment of recurrent pregnancy loss. Am J Reprod Immunol 34: 333– 337, 1995

200. Vaquero E, Valensise H, Menghini S et al: Pregnancy outcome in recurrent spontaneous abortion associated with antiphospholipid antibodies: A comparative study of intravenous immunoglobulin versus prednisone plus low-dose aspirin. Am J Reprod Immunol Microbiol 45: 174– 179, 2001

201. Harris EN, Pierangeli SS: Utilization of intravenous immunoglobulin therapy to treat recurrent pregnancy loss in the antiphospholipid syndrome:A review. Scand J Rheumatol Suppl 107: 97– 102, 1998

202. Johnson PM, Ramsden GH: Recurrent miscarriage. Baillieres Clin Immunol Allergy 2: 607– 624, 1992

203. Thaler CJ: Immunologic role for seminal plasma in insemination and pregnancy. Am J Reprod Immunol 21: 147– 150, 1989

204. Coulam CB, Stern JJ: Seminal plasma treatment of recurrent spontaneous abortion. In Dondero F, Johnson PM (eds): Reproductive Immunology. Serono Symposia 97, pp 205–216. New York, Raven Press, 1993

205. Agrawal S, Pandey MK, Pandey A: Prevalence of MLR blocking antibodies before and after immunotherapy. J Hematother Stem Cell Res 9: 257– 262, 2000

206. Prigoshin N, Tambutti ML, Redal MA et al: Microchimerism and blocking activity in women with recurrent spontaneous abortion (RSA) after alloimmunization with the partner's lymphocytes. J Reprod Immunol 44: 41– 54, 1999

207. Ito K, Tanaka T, Tsutsumi N et al: Possible mechanisms of immunotherapy for maintaining pregnancy in recurrent spontaneous aborters: Analysis of anti-idiotypic antibodies directed against autologous T-cell receptors. Hum Reprod 14: 650– 655, 1999

208. Gafter U, Sredni B, Segal J, Kalechman Y: Suppressed cell-mediated immunity and monocyte and natural killer cell activity following allogenic immunization of women with spontaneous recurrent abortion. J Clin Immunol 17: 408– 419, 1997

209. Fraser EJ, Grimes DA, Schulz KF: Immunization as therapy for recurrent spontaneous abortion: A review and meta-analysis. Obstet Gynecol 82: 854– 859, 1993

210. The Recurrent Miscarriage Immunotherapy Trialist Group: Worldwide collaborative observational study and meta-analysis of allogenic leukocyte immunotherapy for recurrent spontaneous abortion. Am J Reprod Immunol 32:55–72, 1994

211. Hill JA, Anderson DJ: Blood transfusions for recurrent abortion? Is the treatment worse than the disease? Fertil Steril 46: 152– 153, 1986

212. Hormeyr GJ, Jaffe MI, Bezwoda WR, VanIdekinge B: Immunologic investigation of recurrent pregnancy loss and consequences of immunization with husbands leukocytes. Fertil Steril 48: 681– 684, 1987

213. Katz I, Fisch B, Amit S et al: Cutaneous graft-versus host-like reaction after paternal lymphocyte immunization for prevention of recurrent abortion. Fertil Steril 57: 927– 929, 1992

214. Christiansen OB, Mathiesen O, Husth M et al: Placebo controlled trial of active immunization with third-party leukocytes in recurrent miscarriage. Acta Obstet Gynecol Scand 73: 261– 268, 1994

215. Mowbray JF, Gibbings C, Liddell H et al: Controlled trial of treatment of recurrent spontaneous abortion by immunostimulation with paternal cells. Lancet 1: 941– 943, 1985

216. Daya S, Gunby J: The effectiveness of allogeneic leukocyte immunization in unexplained primary recurrent spontaneous abortion. Recurrent Miscarriage Immunotherapy Trialists Group. Am J Reprod Immunol 32: 294– 302, 1994

217. Ober C, Karrison T, Odem RR et al: Mononuclear-cell immunization in prevention of recurrent miscarriages: A randomized trial. Lancet 354: 365– 369, 1999

218. Rodger C: Lack of a requirement for a maternal humoral immune response to establish and maintain successful allogenic pregnancy. Transplantation 40: 372– 375, 1985

219. Ossa JE, Cadavid AP, Maldonado JG: Is the immune system necessary for placental reproduction? A hypothesis on the mechanisms of alloimmunotherapy in recurrent spontaneous abortion. Med Hypotheses 42: 193– 197, 1994

220. Croy BA: The application of SCID mouse technology to questions in reproductive biology. Lab Anim Sci 43: 123– 126, 1993

221. Regan L, Braude PR, Trembath PL: Influence of past reproductive performance on risk of spontaneous abortion. Br Med J 299: 541– 545, 1989

Back to Top