platelet and coagulation - uw hematology protocolswilliams.medicine.wisc.edu/hivcoag.pdf ·...

13
389 © 2002 Schattauer GmbH, Stuttgart Thromb Haemost 2002; 88: 389– 401 Platelet and Coagulation Defects Associated with HIV-1-Infection Simon Karpatkin, Michael Nardi, David Green New York University School of Medicine, New York, USA Platelet Defects HIV-1 seropositive individuals: homosexuals (1), intravenous nar- cotic addicts (2), hemophiliacs (3), and their heterosexual partners (4) develop chronic immunologic thrombocytopenic purpura (ITP). The thrombocytopenia can present early in the course of HIV-1-infection as well as in association with AIDS. The early onset disease (emphasized in this review) is clinically indistinguishable from classic autoimmune thrombocytopenic purpura (ATP) seen predominantly in females, with respect to increased megakaryocytes in the bone marrow, peripheral destruction of antibody-coated platelets and/or impaired platelet production, negative antinuclear antibody, and response to prednisone, i. v. globulin and/or splenectomy. The disease is different from classic ATP with respect to the markedly increased platelet-associated IgG, C3C4, the presence of circulating immune complexes and the male predominance. Historical Descriptions Homosexual ITP (HSITP) Shortly after the first recognition of Kaposi’s sarcoma in sexually- active homosexuals at the Bellevue Hospital Oncology Clinic of New York University Medical Center (5), an “epidemic” of thrombocytope- nia was recognized in the same cohort of patients at the same instituti- on in November 1980. The syndrome was clinically indistinguishable from classic ATP, which is seen predominantly in females (11). Eleven severe cases were first described with a mean platelet count of 16,000 ± 3,000/L, as well as two mild cases. All patients had increa- sed megakaryocytes in the bone marrow, no splenomegaly, negative antinuclear antibody titers, and no clinical disorders known to cause thrombocytopenia. Other findings included: decreased helper/suppres- sor T cell ratios, elevated platelet bound IgG, elevated circulating immune complexes, relative lymphopenia and elevated globulin levels. Both the patient group as well as a control homosexual group (normal complete blood count) had similar increased exposure to her- pes simplex virus, cytomegalovirus, Epstein-Barr virus (EBV), hepati- tis A and B viruses; and similar histories for the use of recreational drugs: marijuana, amyl nitrate, and cocaine. HLA-typing for A-, B-, C- and D-loci revealed no significant associations. Narcotic Addict ITP (NITP) In November 1982 (2 years later), a similar epidemic of chronic ITP was noted in 70 intravenous (IV) narcotic addicts treated at Bellevue Hospital (female to male ratio: 1 to 3), with mean platelet counts of 53,000 ± 4,000 (range 13,000 to 140,000/L) (2). These patients were or had been chronic intravenous abusers of heroin, cocaine, or both for a mean duration of 10 years. Thirty-three patients had stopped taking IV drugs for an average of 21 months, indicating that the thrombocyto- penia was not caused by acute exposure to narcotics. Other possible causes of thrombocytopenia such as classic ATP, hypersplenism, chronic active hepatitis, or AIDS had been ruled out. Elevated platelet bound IgG, C3C4 and serum circulating immune complexes were also noted. Hemophiliac ITP (HITP) In 1983, Ratnoff et al. (3) reported on the development of ITP in five patients with hemophilia (AHF deficiency) who were multi- ply-transfused with lyophilized AHF concentrates for 5 to 9 years with a mean platelet count of 34,600 ± 29,000 (range 8,000 to 82,000/L). This syndrome was clinically indistinguishable from classic ATP. All five had elevated platelet-bound IgG levels. These patients also presented with hypergammaglobulinemia, elevated circulating immune complexes, lymphopenia, decreased helper/suppressor T-cell ratios and positive serology for HIV-1. Heterosexually Transmitted HIV-1-Related ITP In 1987 six cases of HIV-1-related ITP were recognized in “non- risk” individuals who had acquired the disorder through heterosexual contact (4). One such patient was a 64-year-old white woman whose contact was her husband who had received an HIV-1-contaminated blood transfusion for a coronary bypass. The patient developed thrombocytopenia approximately 2 years after resumption of sexual intercourse. Thus, HIV-1-related ITP can be disguised as classic ATP, and is therefore part of the differential diagnosis of unexplained thrombocytopenia. A careful social-sexual history is mandatory for diagnosing patients. Thrombotic Thrombocytopenic Purpura (TTP) In 1988, eight cases of TTP associated with HIV-1 infection were reported (6-8). All 8 cases had been recognized since 1985, relatively late in the development of the HIV-1 epidemic. Five cases reported from NYU Medical Center (7) from 1985 to 1987 represented one third of all cases of TTP diagnosed at the institution. When this rate of inci- Correspondence to: S. Karpatkin, New York University School of Medici- ne, 550 First Avenue, New York, N.Y. 10016, USA – Tel.: 212 263-5609, Fax: 212 263-0695; E-mail: [email protected] Review Article

Upload: buidiep

Post on 01-Mar-2019

218 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Platelet and Coagulation - UW Hematology Protocolswilliams.medicine.wisc.edu/hivcoag.pdf · Platelet and Coagulation Defects Associated ... and hemolytic uremic syndrome ... An analysis

389

© 2002 Schattauer GmbH, Stuttgart Thromb Haemost 2002; 88: 389–401

Platelet and Coagulation Defects Associated with HIV-1-Infection

Simon Karpatkin, Michael Nardi, David Green

New York University School of Medicine, New York, USA

Platelet Defects

HIV-1 seropositive individuals: homosexuals (1), intravenous nar-cotic addicts (2), hemophiliacs (3), and their heterosexual partners (4)develop chronic immunologic thrombocytopenic purpura (ITP). Thethrombocytopenia can present early in the course of HIV-1-infection aswell as in association with AIDS. The early onset disease (emphasizedin this review) is clinically indistinguishable from classic autoimmunethrombocytopenic purpura (ATP) seen predominantly in females, withrespect to increased megakaryocytes in the bone marrow, peripheral destruction of antibody-coated platelets and/or impaired platelet production, negative antinuclear antibody, and response to prednisone,i.v. � globulin and/or splenectomy. The disease is different fromclassic ATP with respect to the markedly increased platelet-associatedIgG, C3C4, the presence of circulating immune complexes and the male predominance.

Historical Descriptions

Homosexual ITP (HSITP)

Shortly after the first recognition of Kaposi’s sarcoma in sexually-active homosexuals at the Bellevue Hospital Oncology Clinic of NewYork University Medical Center (5), an “epidemic” of thrombocytope-nia was recognized in the same cohort of patients at the same instituti-on in November 1980. The syndrome was clinically indistinguishablefrom classic ATP, which is seen predominantly in females (11). Elevensevere cases were first described with a mean platelet count of 16,000 ± 3,000/�L, as well as two mild cases. All patients had increa-sed megakaryocytes in the bone marrow, no splenomegaly, negativeantinuclear antibody titers, and no clinical disorders known to causethrombocytopenia. Other findings included: decreased helper/suppres-sor T cell ratios, elevated platelet bound IgG, elevated circulating immune complexes, relative lymphopenia and elevated � globulin levels. Both the patient group as well as a control homosexual group(normal complete blood count) had similar increased exposure to her-pes simplex virus, cytomegalovirus, Epstein-Barr virus (EBV), hepati-tis A and B viruses; and similar histories for the use of recreationaldrugs: marijuana, amyl nitrate, and cocaine. HLA-typing for A-, B-, C-and D-loci revealed no significant associations.

Narcotic Addict ITP (NITP)

In November 1982 (2 years later), a similar epidemic of chronic ITPwas noted in 70 intravenous (IV) narcotic addicts treated at BellevueHospital (female to male ratio: 1 to 3), with mean platelet counts of53,000 ± 4,000 (range 13,000 to 140,000/�L) (2). These patients wereor had been chronic intravenous abusers of heroin, cocaine, or both fora mean duration of 10 years. Thirty-three patients had stopped takingIV drugs for an average of 21 months, indicating that the thrombocyto-penia was not caused by acute exposure to narcotics. Other possiblecauses of thrombocytopenia such as classic ATP, hypersplenism, chronic active hepatitis, or AIDS had been ruled out. Elevated plateletbound IgG, C3C4 and serum circulating immune complexes were alsonoted.

Hemophiliac ITP (HITP)

In 1983, Ratnoff et al. (3) reported on the development of ITP in five patients with hemophilia (AHF deficiency) who were multi-ply-transfused with lyophilized AHF concentrates for 5 to 9 years witha mean platelet count of 34,600 ± 29,000 (range 8,000 to 82,000/�L).This syndrome was clinically indistinguishable from classic ATP. Allfive had elevated platelet-bound IgG levels. These patients also presented with hypergammaglobulinemia, elevated circulating immunecomplexes, lymphopenia, decreased helper/suppressor T-cell ratios andpositive serology for HIV-1.

Heterosexually Transmitted HIV-1-Related ITP

In 1987 six cases of HIV-1-related ITP were recognized in “non-risk” individuals who had acquired the disorder through heterosexualcontact (4). One such patient was a 64-year-old white woman whosecontact was her husband who had received an HIV-1-contaminatedblood transfusion for a coronary bypass. The patient developed thrombocytopenia approximately 2 years after resumption of sexual intercourse. Thus, HIV-1-related ITP can be disguised as classic ATP,and is therefore part of the differential diagnosis of unexplained thrombocytopenia. A careful social-sexual history is mandatory for diagnosing patients.

Thrombotic Thrombocytopenic Purpura (TTP)

In 1988, eight cases of TTP associated with HIV-1 infection were reported (6-8). All 8 cases had been recognized since 1985, relativelylate in the development of the HIV-1 epidemic. Five cases reportedfrom NYU Medical Center (7) from 1985 to 1987 represented one thirdof all cases of TTP diagnosed at the institution. When this rate of inci-

Correspondence to: S. Karpatkin, New York University School of Medici-ne, 550 First Avenue, New York, N.Y. 10016, USA – Tel.: 212 263-5609, Fax: 212 263-0695; E-mail: [email protected]

Review Article

Page 2: Platelet and Coagulation - UW Hematology Protocolswilliams.medicine.wisc.edu/hivcoag.pdf · Platelet and Coagulation Defects Associated ... and hemolytic uremic syndrome ... An analysis

390

Thromb Haemost 2002; 88: 389–401

dence, 5 of 15 patients, was compared with the incidence of patientshospitalized with AIDS during the same period (3560 of 171,210), thedifference was significant by chi square analysis, indicating that the apparent association is not caused by ascertainment bias.

Thrombotic microangiopathy (TMA) is a heterogeneous disorderthat includes both classic thrombotic thrombocytopenic purpura (TTP)and hemolytic uremic syndrome (HUS). Intravascular platelet aggrega-tes are characteristic of the pathologic lesions with sparing of the liverand lung (9) and von Willebrand factor expression is a prominent feature (10). TMA has been subsequently described in over 150 HIV-1cases (6, 7, 11-14).

Incidence and Prognosis

HIV-1-ITP. In the early description of the epidemic, sexually-activehomosexual patients presented with ITP in the presence as well as absence of AIDS. Although many patients developed ITP early afterHIV-1 infection, at least 11 other ITP patients had not developed AIDS4-7.5 years (mean 5.2 years) after HIV-1 seropositivity (15). Severalpatients studied (9 patients) reverted to normal platelet counts 5-27months (median 10 months) after the diagnosis of thrombocytopenia in the absence of treatment (i.e. splenectomy or steroids). One of thesepatients presented with a platelet count of 11,000/�L.

Thrombocytopenia can occur without clinical symptoms and increa-ses in incidence with duration of illness and the development of AIDS.For example, the incidence of thrombocytopenia in an asymptomaticgroup of 26 seropositive homosexual men was 0 percent in 1987 (16).This incidence rose to 8 percent in 59 patients with non-AIDS HIV-1-related symptoms and 30 percent in 20 patients with AIDS. Theincidence of thrombocytopenia (<100,000/�L) in 679 HIV-infected i.v. drug abusers was 21-37% (17, 18); 5% with platelet counts< 30,000/�L (18). The incidence of thrombocytopenia in 1711 HIV-1-infected patients with hemophilia varied from 5-19% (19, 20). The incidence rose in the first 5 years, with the risk increasing to 50% in older patients 1 year after the development of AIDS (20).

An analysis of 44 HIV-1-ITP patients studied for a period of 844person-months revealed no greater risk for the development of AIDSthan a similar non-thrombocytopenic seropositive cohort over a periodof 36 months (21).

HIV-1-TTP. While complete remissions are observed with appro-priate treatment in HIV-associated TTP, unlike classic TTP the repor-ted long-term survival is poor, with no survivors reported beyond 2 years (11, 13). HIV-associated TTP may differ from classic TTP in itsgradual onset, presence of concurrent medical illness, occasional spon-taneous improvement and inferior response to treatment (11).

Treatment

The rationale for treatment of HIV-1-ITP has been to use the lowestpossible corticosteroid dose capable of raising the platelet count to safelevels, generally > 25,000/�L with absence of significant purpura.Prednisone is administered at a dose of 30 to 40 mg/day for 1 to 2 weeksand rapidly tapered to a maintenance dose of 10 to 15 mg/day. The patient is then observed for 10 months, if possible, which is the mediantime for spontaneous reversion to a normal platelet count in 18% of ourpatients studied. If the patient has not obtained a safe platelet count bythis time, or requires more than 10 to 15 mg of prednisone per day tomaintain such a platelet count, splenectomy is performed. In our studyof 41 homosexual patients, 35 had a moderate (>50,000/�L) to excel-lent (>100,000/�L) response while on corticosteroids (30 relapsed

following cessation of corticosteroids). An excellent response tosplenectomy was noted in 27 of 32 patients (84%) with a mean follow-up time of 14 months (15, 22, 23). Of our original cohort of homosexualpatients, 26 of 41 (63%) did not require treatment for their throm-bocytopenia. There is no rigorous evidence that corticosteroid treat-ment or splenectomy contributes to the development of AIDS. Predni-sone therapy has been complicated, however, by the development oforal candidiasis as well as activation of latent herpes simplex virus.

The efficacy of azidodeoxythymidine (AZT) in the treatment of ITPin homosexuals is often dramatic (24, 25). There is an exponential risein platelet count within the first 1 to 2 weeks of AZT treatment (200 mgevery 6 h), with the maintenance of normal or higher counts for 7 weeks(duration of study). Hematocrits decreased in an exponential mannerduring the same period of time. After discontinuation of AZT, the platelet count remained elevated for more than 4 weeks in 3 of 5 pa-tients. These observations on the relatively rapid rise in platelet countfollowing AZT treatment may therefore suggest that HIV-1 infection ofbone marrow precursor cells, megakaryocytes or stromal cells may beinhibiting platelet production (see below). Similar results have been reported with the new anti-retroviral drugs: Indinavir, Saquinavir andritonavir. Twenty-three patients treated without AZT increased theirplatelet counts from 63,000 to 125,000/�l and decreased their viral load 74 fold during 6 months of observation with this treatment. Nocorrelation was noted with CD4 count (26).

Intravenous gamma globulin (1 to 2 g/kg for 2 to 5 days) is usuallyeffective in the treatment of HIV-related thrombocytopenia; however,its duration is transient, 7 to 10 days (27-30). Six of eight homosexualpatients had a significant rise in their platelet count, as did all of 3 drugabusers and all of eight hemophiliacs (27). Another group reported agood to excellent initial response in 12 of 17 patients (71%) (28). Pa-tients refractory to prednisone will occasionally respond to gamma globulin given with steroids.

Intravenous anti-D treatment for Rh+ nonsplenectomized patients isalso effective in the treatment of the disease. HIV-1-infected childrenhave been reported to have the best results with an effect lasting 21 daysin 50% of the responders (31).

Mechanisms of Thrombocytopenia

The mechanism of thrombocytopenia in patients with homosexual,i. v. narcotic addict, and hemophiliac chronic ITP (HSITP, NITP, andHITP, respectively) have been studied extensively by our group (1, 2,32-39) in patients with early onset HIV-1-infection in the absence ofAIDS where the mechanism appears to be increased platelet destruction(40) and compared with those of classic ATP. Myelodysplastic featureswith hypocellularity have been reported in patients with long-termHIV-infection and hemophilia (median 12.5 yrs from seroconversion)(41). In another study in children, TPO levels were 5 fold elevated insevere thrombocytopenic patients who did not respond to i.v. gammaglobulin suggesting marrow failure (42).

Increased Platelet Destruction

Autologous platelet survival studies performed in 13 HSITP patientswith a mean platelet count of <33,000/�L demonstrated a platelet survival of less than 1 day (normal survival time 8 to 10 days). Plateletsequestration was exclusively to predominantly splenic in 85 percent ofthe patients studied. Calculated blood flow and mean transit time werenormal, ruling out hypersplenism (43). Autologous platelet survivalstudies performed in 19 i.v. drug abuser HIV-1-ITP patients with a

Page 3: Platelet and Coagulation - UW Hematology Protocolswilliams.medicine.wisc.edu/hivcoag.pdf · Platelet and Coagulation Defects Associated ... and hemolytic uremic syndrome ... An analysis

391

Karpatkin et al.: Platelet and Coagulation Defects Associated with HIV-1-Infection

mean platelet count of <31,000/�L were < 1 day with no evidence ofsplenic sequestration (43). In another study mean platelet lifespan in 7 patients was 3 ± 4 h with a marked increase in platelet production butinsufficient to maintain a normal platelet count. Five patients with nor-mal platelet counts had a compensated thrombocytolytic state (44). Theabove data support an increased peripheral platelet destruction mechanism for the pathophysiology. Indeed the rapid improvement following treatment with steroids and/or splenectomy in most early-onset HIV-1-infected patients is strongly suggestive of this pattern. However, others have reported impaired platelet production, as well asdecreased platelet survival (see below).

Decreased Platelet Production

Kinetic data on the rapid rise in platelet count (as early as 1 week)following AZT administration suggested that impaired megakaryocyteproduction, platelet release, or both may also be involved (24, 25). Therapid rise in platelet count following the use of AZT precludes a priorchange in anti-platelet Ab levels and suggests that this drug eitherblocks reticuloendothelial function or prevents HIV-1-induced damageof megakaryocytes, which otherwise results in ineffective thrombo-poiesis; or stimulates platelet production. Three platelet kinetic reportssuggest that impaired thrombopoiesis may be operative in patients withHSITP. In one study, platelet production was normal or decreased in 7of 14 patients despite a decreased platelet survival (45). Another groupstudied platelet survival and turnover in 19 homosexual patients withITP treated with and without AZT (46). Both cohorts had a decreasedplatelet survival of about 50 percent of normal. Platelet turnover (pro-duction) was significantly decreased (approximately half the normal rate) in the untreated group (13 patients), compared with normal or increased turnover in the treated group (6 patients). Two patients werestudied before and after AZT administration. Their rise in platelet countwas associated with a three-to-six-fold rise in platelet turnover, with nochange in platelet survival. Thus, AZT appears to exert its effect by enhancing platelet production, suggesting that the thrombocytopeniamay also be related to impairment of the production or function of megakaryocytes. A third study of 6 patients revealed a combination ofshortened autologous platelet survival by 2/3 normal, doubling of sple-nic platelet sequestration and ineffective delivery of platelets to the pe-ripheral blood despite a 6 fold increase in TPO and a 3 fold increase inmegakaryocyte mass (47). These observations are supported by a studyof hemopoietic progenitor cells in 15 patients with AIDS or HIV-1 in-fection, 3 of whom were thrombocytopenic (48). These investigatorsreported a significant impairment of growth of bone marrow mega-karyocyte colony-forming units, as well as granulocyte, erythocyte, andmacrophage precursors in comparison with normal controls, which wascorrected by depletion of T cells and reversed by readdition of autolo-gous T cells. Because most patients have normal to increased mega-karyocytes in their marrow, it is likely that the impairment is caused byineffective thrombopoiesis (platelet production and release). The me-chanism of inhibition of thrombopoiesis is currently unresolved.

An unlikely possibility is the direct infection of megakaryocyteswith HIV-1 (49, 50). Megakaryocytes have CD4 (51, 52) as well asCXCR4 (53, 54), known receptors for HIV-1gp120 and various mega-karyocyte lines are infectable with HIV-1 (52, 55). However reports ofHIV-1 infection of wild-type human megakaryocytes reveal little to noinfectivity (1-5% of cells) compared to T cells or macrophages withquestionable clinical significance (49).

There is now abundant evidence from several laboratories thatCD34+ stem/precursor hematopoietic cells are incapable of being infec-

ted in vivo or in vitro with HIV-1 (56-65). It is therefore logical to suggest that HIV-1-infected stromal cells may be responsible for the well-documented impaired megakaryocytopoiesis of HIV-1-infected patients (48, 58, 65, 66). This could be via induction of inhibitory cyto-kines. This is supported by impaired CFU-MK in 15 HIV-1-seropositivepatients compared to control bone marrows (47, 48), the morphologicalabnormalities detected in the MK of HIV-1-ITP patients (67) decreasedMK precursors (48, 58, 65, 66), and impaired MK survival due to in-creased apoptosis (66). Several cytokines have been shown to inhibitMk growth and differentiation: TGF�-1 (68-72), PF-4 (73), TNF� (74,75), � and � IFN (76) and IL-4 (77). Of interest is the observation thatHIV-1 tat protein, released by infected cells stimulate macrophage pro-duction of TGF�-1 (72), one of the most potent negative regulators ofhematopoiesis, resulting in a dose-dependent inhibition of CFU-MK. Analternative and/or additional mechanism could be autoimmune destruc-tion of MK’s or their precursor by anti-platelet/MK Ab.

These apparently contradictory conclusions regarding decreasedproduction vs increased destruction of platelets can be reconciled,perhaps by the observations of Najean and coworkers who performedplatelet kinetic studies on HIV-1-ITP patients with and without AIDS.They concluded that patients with AIDS are more likely to have decrea-sed platelet production, whereas patients with early onset HIV-1 infec-tion are more likely to have increased peripheral destruction of platelets(40).

Increased Platelet Consumption

TTP is associated with an absent VWF-cleaving Zn metalloproteaseof the ADAMTS13 family (a disintegrin and metalloprotease) in the familial form (78) and the presence of an IgG inhibitor of the proteasein the non-familial form (79, 80). In HUS (81) and in bone marrowtransplant associated TMA normal protease levels are detected (82).Absent VWF-cleaving protease and associated inhibitor have also beendocumented in a case report of HIV-associated TTP (83). The availa-bility of a standardized VWF-cleaving protease assay may allow forimproved classification and management of TMA and elucidate theprecise role of HIV in pathogenesis. In light of its proposed mechanism,TTP may be viewed similarly to ITP, as a manifestation of the clinicalspectrum of HIV-related autoimmunity.

Immunologic Measurements of Platelet Antibody and Immune Complexes

Immunologic measurements of platelet associated IgG, C3C4 andserum circulating immune complexes (CIC), determined by the PEGmethod in homosexual ITP (HSITP), narcotic addict ITP (NITP) andhemophiliac ITP (HITP) reveal strikingly elevated values compared tothe values found in classic ATP patients, i.e. 2.6-3.8 fold greater IgG,2.2-3.9 fold greater C3C4, 2.4-7.3 fold greater PEG-IC. Intermediateelevations of these measurements were noted in non-thrombocytopenicHIV-1-seropositive individuals of the same cohorts (2, 32, 38). Thesedata suggested that circulating immune complexes may be responsiblefor the elevated platelet levels and that no relationship existed betweenthe platelet count and the platelet associated IgG or C3C4. This provedto be the case. Isolated CIC were shown to bind to normal platelets in aconcentration-dependent manner; indeed the level of serum CIC corre-lated with platelet bound IgG, r = 0.5, p <0.05, n = 17. However, un-like the situation with classic ATP, there was no inverse relationship between platelet count and platelet associated IgG, r = -0.2, p >0.1, n = 16 (similar results were reported by others); nor was the platelet

Page 4: Platelet and Coagulation - UW Hematology Protocolswilliams.medicine.wisc.edu/hivcoag.pdf · Platelet and Coagulation Defects Associated ... and hemolytic uremic syndrome ... An analysis

392

Thromb Haemost 2002; 88: 389–401

count inversely related to CIC’s, r = –0.2, p >0.1, n = 20. There-fore, although the platelet immunologic/serum CIC profile is of valuediagnostically in differentiating the thrombocytopenia from classic ATP,it is of no value in predicting platelet count or response to therapy.

Serum anti-platelet binding has been reported employing an indirectsemiquantitative platelet suppression immunofluorescence test in ho-mosexual patients, with impaired binding to Glanzmann’s throm-basthenia platelets [devoid of platelet GPIIb/IIIa (84)]. However, theseauthors did not prove F(ab’)2 binding. A second group, although unableto elute anti-platelet Ab from HIV-1-ITP narcotic addict patients wereable to detect plasma anti-platelet Ib/IX and IIb/IIIa Ab utilizing theMAIPA technique on normal platelets preincubated with patient plasma in 43% of 45 patients (85). Our group has been unable to detectserum F(ab’)2 binding in the homosexual cohort of HIV-1-ITP patients.However we have detected F(ab’)2 binding in HIV-1 seropositive i.v.drug abusers and hemophiliacs (2, 32). The reasons for these differen-ces are likely to be the presence of anti-idiotype blocking Ab (see below).

Composition of Immune Complexes

Anti-F(ab’)2 Antibodies

Studies designed to analyze platelets and immune complexes for viral antigens suggested the presence of anti-antibodies. Fixed washedplatelets or eluates of HSITP patients revealed absence of EBV, cyto-megalovirus (CMV), herpes simplex virus (HSV), or adenovirus(ADE) viral antigens on their platelets, despite the presence of viral an-tibodies for these antigens in their sera. Similar findings were noted infixed immune complexes of these patients. Viral antibodies againstEBV, CMV, HSV, and rubeola virus were noted in these complexes,and correlated with their presence in sera. No such correlation was noted with preparations from healthy control subjects whose immunecomplexes were negative in the presence of positive antiviral serum titers (39).

Following this lead, anti-F(ab’)2 antibodies were sought and foundin 9 of 12 HSITP and 6 of 6 i.v. drug abusers, which correlated with im-mune complex levels, r = 0.83, p <0.01, n = 16. In contrast, anti-F(ab’)2

antibodies were not detectable in 6 patients with classic ATP againstF(ab’)2 fragments of subjects from autologous, homologous ATP ornormal control groups (39).

Anti-HIV-1gp120 and Anti-idiotype Antibodies

Analysis of serum PEG-CIC as well as platelet acid eluates of HIV-1-ITP, HSITP and NITP patients has revealed the presence of anti-HIV-1gp120 Ab and its anti-idiotype (33, 86). HIV-1 antigen or provi-ral DNA was not detectable. Approximately 50% of eluted platelet IgGcontained anti-HIV-1gp120 Ab (33).

Anti-GPIIIa Ab In Vitro and In Vivo

PEG-IC’s of HIV-1-ITP patients were purified on protein A col-umns, dissociated in acid, separated by gel filteration on an acidified G200column and affinity-purified on anti-IgG and anti-IgM columns. BothIgG and IgM reactivity was found against HIV-1gp120, HIV-1gp24CD4 receptor, and human platelets. Only the IgM reacted with purifiedFc fragments, indicating rheumatoid factor (RF) reactivity. The IgG an-ti-platelet reactivity was shown to be specific for platelets by demon-strating reactivity with F(ab’)2 fragments (34) (Fig. 1). Anti-plateletIgG reactivity was affinity purified on fixed platelets and the eluate in-cubated with platelet lysates containing 125I-labeled surface plateletmembrane. The immune complex precipitate was then analyzed onSDS-PAGE in the presence and absence of reducing agent. An ~100 Kdpredominant band was noted, which paradoxically increased its mwupon reduction demonstrating the physical characteristics of plateletGPIIIa. This was proven with specific MoAb’s against platelet GPIIIa,simultaneously incubated with platelet lysate (Fig 2) (34). Further stu-dies revealed specificity for amino acids 49-66 on GPIIIa, with an inverse curvilinear relationship between patient anti-platelet IgG serum(as well as anti-GPIIIa49-66) and platelet count (Fig. 3). The in vivo pathophysiologic relevance of these in vitro observations was substan-tiated by demonstrating that ~25 �g of affinity-purified anti-plateletIgG1 of HIV-1-ITP PEG-IC’s induced dramatic thrombocytopenia inrecipient mice (which have 83% homology with human GPIIIa and Fc receptors for human IgG1). In vivo specificity was substantiated by demonstrating prevention or reversal of the anti-GPIIIa-inducedthrombocytopenia with an albumin conjugate of GPIIIa49-66 (Fig. 4)(37).

The presence of this immunodominant autoimmune GPIIIa49-66epitope appears to be unique, since classic ATP appears to have multi-ple epitopes within platelet membrane receptors, GPIIb/IIIa and GPIb.

Fig. 1 Anti-platelet reactivity of disso-ciated IgG and IgM of immune complexescompared to HIV-1-ITP patient serumIgG. Washed platelets (1�107) were ap-plied to plastic microtiter wells in PBS for1 h at room temperature, blocked withPBS/1% BSA, and then assayed by ELISAfor IgG or IgM. (A) Anti-platelet reactivityof dissociated IgG and serum IgM. (B) An-ti-platelet reactivity of dissociated F(ab’)2

fragments of IgG vs. serum IgG (n = 4)

Page 5: Platelet and Coagulation - UW Hematology Protocolswilliams.medicine.wisc.edu/hivcoag.pdf · Platelet and Coagulation Defects Associated ... and hemolytic uremic syndrome ... An analysis

393

Karpatkin et al.: Platelet and Coagulation Defects Associated with HIV-1-Infection

The observations in HIV-1-ITP could be related to cross-reactivity withHIV-1 antigens. This is suggested from the work of Bettaieb et al whonoted cross-reactivity with HIV-1gp120 and platelet GPIIIa (87); ourlaboratory which found anti-HIV-1gp120 on platelets of HIV-1-ITP patients (33); Gonzalez-Conejero et al. who noted cross-reactivity withHIV-1gp120 of serum-platelet reactive eluates with GPIIb/IIIa andGPIb/IX in 20% of 45 drug abusers (85); and Chia et al. (88) who described cross-reactive HIV-1p24 and HIV-1gp120 Ab’s in alkalineand acid eluates of serum Ab’s bound to an HIV sepharose 4B affinitycolumn. Indeed 48% of alkaline-eluted Ab bound to platelets as well asp24.

Rheumatoid Factor Antibodies

Elevated CD5+ B cells have been reported in patients and mice withautoimmune diseases as well as in HIV-1-ITP (89). CD5+ B cells gene-rally produce low affinity antibodies, which are predominantly RF ofthe IgM class against Fc of IgG (90). Because PEG-IC’s of these pati-ents contain high concentrations of IgM, RF-IgG complexes weresought within their serum PEG-IC’s. Purified PEG-IC IgM was shownto react with purified PEG-IC IgG of these patients, relocating ~50% ofthe IgG preincubated with IgM to the Vo (void volume) region of aG200 gel filtration column (34). Thus PEG-IC’s of HIV-1-ITP patients

Fig. 2 Immunoprecipitation of 125I-platelet surface antigens with anti-platelet IgG. (A) Experiment 1. Lanes 1 and 2 contain platelet lysates of RM (homosexualITP) treated with anti-platelet IgG, unreduced and reduced, respectively; lanes 3 and 4 were treated with control IgG, unreduced and reduced, respectively. (B) Ex-periment 2. Lanes 1, 3 and 5 are unreduced. Lanes 2, 4 and 6 are reduced. Lanes 1 and 2, anti-platelet IgG of MM (intravenous drug abuser ITP); lanes 3 and 4,treated with monoclonal antibody LK3r against GPIIIa; lanes 5 and 6, treated with monoclonal antibody LK7r against GPIIIa. Control IgG (not shown) gave noprecipitation as in experiment 1, lanes 3 and 4. Arrow, 100-kDa band

Fig. 3 Correlation of platelet count with anti-platelet IgG and anti-GPIIIa-(49-66). (A) Anti-platelet IgG was extracted from PEG-ICs, affinity-purified with fi-xed platelets, and tested for its reactivity with washed platelets adsorbed to a microtiter plate via a dilution determination of antibody titer, as described. A best-fitcurvilinear power regression curve was selected by computer analysis (Macintosh DELTA GRAPH PRO 3.5) with R2 = 0.61, F(1,25) = 40.2, and P< 0.0001. (B)Anti-GPIIIa-(49-66) concentration was measured in serum by using a standard curve of affinity-purified anti-GPIIIa-(49-66) vs. purified peptide. Ordinate repre-sents nanograms of antibody per ml of sera, diluted 1:4, reacting with GPIIIa-(49-66) adsorbed to microtiter plates. Gray horizontal band represents mean ± 2 SDof 10 control subjects tested. A similar best-fit curvilinear power regression curve was selected with R2 = 0.51, F(1,29) = 30.0, and P< 0.0001

A B

PLATELET COUNT/µl � 103 PLATELET COUNT/µl � 103

Page 6: Platelet and Coagulation - UW Hematology Protocolswilliams.medicine.wisc.edu/hivcoag.pdf · Platelet and Coagulation Defects Associated ... and hemolytic uremic syndrome ... An analysis

394

Thromb Haemost 2002; 88: 389–401

Fig. 4 In vivo induction of thrombocytopenia in mice injected with affinity-purified anti-platelet IgG of HIV-1-ITP patients and its reversal with GPIIIa-(49-66).(A) Thirty micrograms of anti-platelet IgG of patients 6 and 11 or purified control human IgG was injected i.p. into eight experimental and six control mice, respectively. (B) Twelve mice were injected i.p. with 25 �g of anti-platelet IgG of patient 11, as above. Seven of these mice were simultaneously injected in theopposite flank with 226 nmol of GPIIIa-(49-66)-albumin conjugate, whereas five were simultaneously injected with the irrelevant scrambled GPIIIa-albumin conjugate (CGGGARVLEDRP) at the same concentration. (C) Ten mice were injected i.p. with 50 �g of anti-platelet IgG of patients 10 and 11, as above. At 2 h, six of these mice were injected with GPIIIa-(49-66)-albumin conjugate, whereas four were injected with the irrelevant-scrambled GPIIIa-albumin conju-gate

A

C

B

Page 7: Platelet and Coagulation - UW Hematology Protocolswilliams.medicine.wisc.edu/hivcoag.pdf · Platelet and Coagulation Defects Associated ... and hemolytic uremic syndrome ... An analysis

395

Karpatkin et al.: Platelet and Coagulation Defects Associated with HIV-1-Infection

contain RF-IgG complexes as well as anti-HIV-1gp120-anti-idiotypecomplexes.

Anti-GPIIIa Ab and Anti-idiotype Ab

Serum from HSITP and NITP patients have little anti-platelet Ab aswell as anti-GPIIIa49-66 Ab compared to purified serum IgG sug-gesting the presence of blocking Ab. Indeed no inverse correlation wasnoted between platelet associated IgG and platelet count. This appearedto be different from our findings in hemophiliac HIV-1 seropositivethrombocytopenic patients (HITP) as well as 15 classic ATP patients,in which serum anti-platelet Ab as well as F(ab’)2 fragment binding waseasily detectable. Indeed 6/6 HITP patients had elutable anti-plateletAb from their platelets and an inverse relationship was noted betweenplatelet count and platelet bount IgG, r = -0.8, p <0.001, n = 28 (34). Similar results were obtained with classic ATP patients, similarly studied (34). To investigate this difference in detectable serum anti-GPIIIa49-66 Ab in HSITP and NITP patients, crude serum, purified serum IgG and PEG-IC IgG were isolated and studied for anti-GPIIIa49-66 reactivity. This revealed ~150 fold greater Ab reactivity inpurified serum IgG and ~4000 fold greater reactivity in PEG-IC IgG(Fig. 5). This was explained by the presence of anti-idiotype Ab2 (bothIgG and IgM) sequestered within the PEG-IC.

The IgM anti-idiotype was further studied since it is predominantlya blocking Ab, as demonstrated by specificity for F(ab’)2 fragments ofanti-GPIIIa49-66, and inhibition with peptide GPIIIa49-66, not with acontrol peptide. The IgM anti-idiotype Ab is not polyreactive. In agroup of HIV-1-infected patients (non-thrombocytopenic as well asthrombocytopenic) a positive correlation was noted between plateletcount and anti-idiotype IgM (r = 0.7, p = 0.0001, n = 22). The in vivorelevance of the IgM anti-idiotype Ab was tested by inducing throm-bocytopenia (–30% baseline) in mice with anti-GPIIIa49-66. Preincu-bation of the anti-GPIIIa49-66 Ab with the IgM anti-idiotype (molar ratio1:7) improved the platelet count to 80% of normal and throm-bocytopenia could be reversed, 4 h after induction of thrombocytopenia

(Fig. 6) (36). Thus CIC’s of HIV-1-infected patients contain blockingIgM anti-idiotype Ab against anti-GPIIIa which regulates their serumreactivity in vitro and level of thrombocytopenia in vivo.

These observations along with the presence of platelet reactive idiotype-anti-idiotype complexes (i.e., HIV-1gp120) as well as the presence of RF offer an explanation for the markedly elevated immunecomplex/Ig on platelets of HIV-1-ITP patients. They also offer an explanation for the presence of IgG on platelets of HIV-1-infected patients with normal platelet counts – namely the presence of sufficientanti-idiotype Ab to neutralize the thrombocytopenia.

Fig. 5 Anti-platelet GPIIIa49-66 reactivity of serum, serum IgG, and IC-IgGof one representative HIV-1-ITP patient of five patients studied. Various IgGconcentrations of serum, purified serum IgG, and purified IC-IgG were assay-ed for anti-GPIIIa49-66 reactivity by solid-phase ELISA

Fig. 6 Prevention and reversal of in vivo thrombocytopenia induced by anti-GPIIIa-(49-66) induced in mice with IgM anti-idiotype Ab against anti-GPIIIa-(49-66). (A) Prevention. Four to seven mice in each group were treated intrape-ritoneally with either anti-GPIIIa-(49-66) Ab, Ab plus control IgM (125 �g).Ab plus anti-idiotype IgM (125 �g). Ab plus anti-idiotype IgM (875 �g), or Abplus anti-idiotype IgG (875 �g). Platelet counts were obtained at various timeintervals for 24 h. The mean platelet count ± SEM for all four groups was similar (1.72 ± 0.56 � 106/�l). Pt, patient. (B) Reversal. 16 mice were treatedintraperitoneally with 25 �g of anti-GPIIIa-(49-66), and 4 mice were treated intraperitoneally with saline. At 4 h, after induction of thrombocytopenia, the16 antibody-treated mice were treated (4 mice in each group) with 875 �g ofeither control IgG, patient (Pt) anti-idiotype IgG, control IgM, or patient anti-idiotype IgM, and were followed for 24 h. Note reversal of thrombocytopeniawith IgM anti-idiotype at 6, 8, and 12 h compared with control IgM (P =0.0008, 0.0015, and 0.0137, respectively; paired Studient’s t test), not with IgGanti-idiotype or control IgG

A

B

Page 8: Platelet and Coagulation - UW Hematology Protocolswilliams.medicine.wisc.edu/hivcoag.pdf · Platelet and Coagulation Defects Associated ... and hemolytic uremic syndrome ... An analysis

396

Thromb Haemost 2002; 88: 389–401

Complement-Independent, Peroxide-induced Anti-GPIIIa49-66 Platelet Lysis

A recent report that recognized the presence of platelet fragmentswithin the serum PEG-IC’s stimulated a search for antibody-dependentplatelet lysis. This was found to occur in vitro with gel-filtered platelets(Fig. 7 and 8) as well as in vivo in association with the induction ofthrombocytopenia. The same results were noted with F(ab’)2 fragmentsas well as intact IgG in complement-deficient mice, indicating the absence of complement-induced lysis. This new pathophysiologic mechanism of platelet destruction was shown to be induced via peroxi-de generation from a platelet-associated NADPH oxidase pathway. Invitro platelet lysis could be blocked by scavengers of reactive oxygenspecies (catalase, superoxide dismutase, diphenyleneniodonium) (Fig. 9) and platelet lysis did not occur in NADPH oxidase-deficientmice (p47phox-/-). Thrombocytopenia was significantly improvedcompared to its induction in wild-type mice by 60% (Fig. 10).

Coagulation Abnormalities

A number of coagulation abnormalities have been described in HIV disease including acquired deficiency states of the physiologic anticoagu-lants: protein C (91), protein S (92) and heparin cofactor II (93). Of these,protein S deficiency is the most consistently observed and is present in upto 73% of HIV-infected men (92, 94, 95). Most commonly there is reduc-tion in free protein S and coordinate reduction in functional protein S activity. C4bBP levels are normal indicating that reduced protein S is not aconsequence of a shift to the bound form, which is typically seen in in-flammatory states. There is no correlation of free protein S deficiency withclinical thrombosis in HIV-infected men (96). Protein S is reduced in numerous other conditions including pregnancy, use of oral contraceptives,DIC, acute thrombosis and liver disease. The clinical significance of acqui-red protein S deficiency associated with HIV disease is unclear. High levelsof plasma von Willebrand factor have been reported in HIV disease andmight be indicative of activated endothelium (97).

Fig. 7 In Vitro Platelet Fragmentation Induced by Anti-GPIIIa49-66 Ab. A. Immunoblot of PEG-IC of control and HIV-1-ITP patients employing anti-GPIIIa,anti-GPIba and control mouse monoclonal Ab, MoPC. CTL refers to control. B. Flow Cytometry of Platelet In Vitro Particle Formation. Gel filtered platelets were labelled with anti-GPIIb-FITC monoclonal Ab, washed and then treated with various agents: buffer, control IgG and patient anti-GPIIIa49-66 for 4 h at 37° C. Three panels represent: CTL, buffer alone; CTL IgG, IgG isolated from control PEG-IC; PT IgG, IgG isolated from HIV-1-ITP patient PEG-IC. X axis refers to fluorescence, Y axis to forward scatter. Numbers in left upper quadrant refer to the % Ab-induced particles with decreased fluorescence attributed to platelet fragmentation. A highly-reactive anti-GPIIIa49-66 IgG is shown. C. Distribution of % platelet particle formation in control vs. HIV-1-ITP vs. ATP patients at 1 h incubation. Results shown for IgG isolated from PEG-IC’s of 12 control, 16 HIV-1-ITP and 5 ATP patients. Box Plot. Mean is shown by the solidblack box; median by the horizontal line in the large open box; 25th and 75th percentiles by the lower and upper border of the large open box from which spread ofthe data from the position of the median can be assessed. Whiskers include 99% of a Gaussian distribution

A

B

C

Page 9: Platelet and Coagulation - UW Hematology Protocolswilliams.medicine.wisc.edu/hivcoag.pdf · Platelet and Coagulation Defects Associated ... and hemolytic uremic syndrome ... An analysis

397

Karpatkin et al.: Platelet and Coagulation Defects Associated with HIV-1-Infection

Anticardiolipin antibodies (aCL) and lupus anticoagulants (LA) arefrequent incidental laboratory findings in HIV-infected patients. Thepresence of LA is an established risk factor for both venous and arteri-al thrombosis in patients with SLE and is detected by a prolonged par-tial thromboplastin time (aPTT) that fails to correct with mixing, or al-ternatively by prolongation of the Kaolin clotting time, diluted tissuethromboplastin time or dilute Russell viper venom time. Elevated aCLare found in 20-70% of HIV patients (98) and are predominantly IgG(99). LA activity is prevalent in HIV-infected patients and consistsmainly of IgM antibody (100, 101). Despite the high prevalence of aCLand LA in HIV-infected patients, the clinical manifestations of the clas-sic antiphospholipid antibody syndrome (APS) such as stroke or TIA,recurrent venous thrombosis, cutaneous lesions and miscarriage or themanifestations of catastrophic APS are distinctly unusual. Only a singlecase report links APS to HIV infection (102). Anti-beta-2-glycoproteinI (-�2GPI) and anti-prothrombin antibodies are seen less commonly inHIV infected patients than in APS/SLE or primary APS patients. Thebiological false positive test for syphilis is rarely detected in HIV-in-

fected patients (103). This antiphospholipid antibody specificity mightexplain the lack of clinically evident thrombosis in the HIV-infectedpopulation. It is suggested that elevated aCL in HIV results from pan-Bcell stimulation/hypergammaglobulinemia or alternatively could resultfrom infection. The presence of aCL and lupus anticoagulant activitydo not correlate with the clinical features of HIV including medicationuse, infections or malignancy or CD4 T-cell count nor with the deve-lopment of thrombosis (96). LAs are not generally considered to pose ableeding risk, however associated thrombocytopenia (104), plateletdysfunction or hypoprothrombinemia (105) may result in unexpectedbleeding particularly after surgery and prophylactic transfusion of pla-telets and plasma may be appropriate in select cases.

HIV and Thrombosis

Venous thromboembolic disease (VTE) is uncommon in AIDS pati-ents but has been described in a number of case reports and in retros-pective reviews (91, 106–108). Thrombosis at unusual sites including

Fig. 8 Electron Microscopy of DamagedPlatelets treated with Anti-GPIIIa49-66 Ab.A: Patient sample at 1 h showing plateletswith a fuzzy material attached to the outersurface of the cell membranes (dotted arro-ws). Gaps are noted in the cell membraneswith leakage of cytoplasmic content (arro-ws). These areas are demonstrated at highermagnification in B and C. E: patient sampleat 4 h showing degenerating platelets and disintegration of the cell membrane (arrow).Swollen platelet (F), platelet fragments (H)and occasional normal platelets (G,I) are alsoseen. None of these changes were present inIgG controls at 4 h (D). Original magnifica-tions: A, D-I: 4,000 X. B: 50,000 X. C:40,000 X

Page 10: Platelet and Coagulation - UW Hematology Protocolswilliams.medicine.wisc.edu/hivcoag.pdf · Platelet and Coagulation Defects Associated ... and hemolytic uremic syndrome ... An analysis

398

Thromb Haemost 2002; 88: 389–401

retinal vein (109) and sagittal sinus (110) have been reported which isalso a characteristic feature of thrombophilic states. The majority ofVTE is associated with advanced HIV (CD4 count <200), underlyingAIDS-related malignancy or opportunistic infection, while idiopathicDVT is less common. It is not known whether the incidence of VTE dif-fers in HIV patients as compared to the general population since thishas not been studied in a prospective case control manner. Given thediagnostic challenge of VTE and the protean clinical manifestations ofHIV disease itself, the incidence of DVT may be underestimated in thispatient population. Further studies will be required to determine thethrombosis risk attributable to HIV and related co-morbidities, as wellas the optimal strategies for DVT treatment and prophylaxis and the utility of thrombophilia screening.

HIV and Hemophilia

AIDS is the leading cause of death in hemophilia. There has not been documented HIV seroconversion since 1986 (111). ITP in HIV-infected hemophiliacs may further worsen the risk of bleeding. The treatment of hemophilia is complicated by the development of high titer factor VIII inhibitors, which may resolve with advanced HIV disease due to loss humoral immunity (112).

A number of reports have suggested that protease inhibitors have thepotential to cause increased bleeding in HIV-infected hemophiliacs(113). Increase in frequency of bleeding episodes, unusual sites of blee-ding and increased factor concentrate requirement have all been noted(114). Ritonavir has received particular attention in this regard, but allthe currently available protease inhibitors are linked to increased blee-ding. The mechanism of bleeding in the majority of cases was not established, although platelet dysfunction was identified as a cause ofbleeding in two hemophiliacs (115). Mucosal bleeding has also been reported in non-hemophiliacs treated with protease inhibitors indicatingthat the effect is not related to factor VIII per se (114, 116).

Fig. 9 Generation of Peroxide and Effect ofPeroxide Inhibitors, Catalase, Diphenylenei-odonium (DPI) and Superoxide Dismutase(SOD) with Platelet Particle Formation Indu-ced by Anti-GPIIIa49-66 Ab or Thrombin. A.Gel filtered platelets were preincubated withinhibitor or buffer for 15 min prior to the ad-dition of rabbit anti-GPIIIa49-66 Ab or con-trol IgG. C refers to control non-immune IgG(similar results with control plus highest inhi-bitor concentration employed), IS refers torabbit anti-GPIIIa49-66 immune serum. Barslabelled 2, 3, 4 after bar 1 refer to doublingconcentrations of inhibitor (50 u/ml catalase,5 nM/ml DPI and 12.5 u/ml SOD), n = 4, SEMis given. B. Generation of peroxide by anti-GPIIIa49-66 at 1 h (panels A, B, C) and 4 h(panels D, E, F). Platelets were loaded withthe intracellular dye, DCFH-DA and thentreated with buffer (panels A and D), controlIgG (panels B and E) or human anti-GPIIIa49-66 (panels C and F). C. Effect of peroxide in-hibitors on thrombin-induced platelet particleformation. Gel-filtered platelets were preincu-bated with inhibitors, as in AA and then treated with 1 u/ml thrombin for 4 h. Bars refer to doubling concentrations, n = 4

Fig. 10 Effect of Anti-GPIIIa49-66 Ab on Platelet Count and Platelet ParticleFormation in Control C57BL/6 and p47phox(-/-) Mice. Control C57BL/6 orp47phox(-/-) mice were injected i.p. with 25 ug of intact anti-GPIIIa49-66 or 16 ug of its F(ab’)2 fragment and platelet count and % platelet particles moni-tored at 2 and 4 h. A. Platelet count. B. % platelet particle. N = 6-8 for eachgroup, SEM is given

A

C

B

A

B

Page 11: Platelet and Coagulation - UW Hematology Protocolswilliams.medicine.wisc.edu/hivcoag.pdf · Platelet and Coagulation Defects Associated ... and hemolytic uremic syndrome ... An analysis

399

Karpatkin et al.: Platelet and Coagulation Defects Associated with HIV-1-Infection

References

1. Morris L, Distenfeld A, Amorosi E, Karpatkin S. Autoimmune throm-bocytopenic purpura in homosexual men. Ann Intern Med 1982; 96: 714-7.

2. Savona S, Nardi MA, Karpatkin S. Thrombocytopenic purpura in narco-tics addicts. Ann Intern Med 1985; 102: 737-41.

3. Ratnoff OD, Menitove JE, Aster RH, Lederman MM. Coincident classichemophilia and “idiopathic” thrombocytopenic purpura in patients undertreatment with concentrates of anti-hemophilic factor (factor VIII). N EnglJ Med 1983; 308: 439-42.

4. Karpatkin S, Nardi M, Hymes KB. Immunologic thrombocytopenic pur-pura after heterosexual transmission of human immunodeficiency virus(HIV). Ann Intern Med 1988; 109: 190-3.

5. Hymes KB, Cheung T, Greene JB, et al. Kaposi's sarcoma in homosexualmen: A report of 8 cases. Lancet 1981; 2: 598-600.

6. Jokela J, Flynn T, Henry K. Thrombotic thrombocytopenic purpura in ahuman immunodeficiency virus (HIV)-seropositive homosexual man. AmJ Hematol 1987; 25: 341-3.

7. Leaf AN, Raphael B, Hochster H, Laubenstein LJ, Baez L, Karpatkin S:Thrombotic thrombocytopenic purpura associated with HIV-1 infection.Ann Intern Med 1988; 109: 194-7.

8. Nair JMG, Bellevue R, Bertoni M, et al. TTP in patients with the AIDS-related complex. Ann Intern Med 1988; 109: 209-12.

9. Gore I. Disseminated arteriolar and capillary platelet thrombosis: A mor-phological study of its histogenesis. Am J Pathol 1950; 26: 155-75.

10. Assada Y, Sumiyoshi A, Hayashi T, Suzumiya J, Kekatani. Thromb Res1985; 38: 469-79.

11. Bell WR, Chulay JD, Feinberg JE. Manifestations resembling thromboticmicroangiopathy in patients with advanced human immunodeficiency virus (HIV) disease in a cytomegalovirus prophylaxis trial (ACTG204).Medicine 1997; 76: 369-80.

12. Hymes KB, Karpatkin S. Human immunodeficiency virus infection andthrombotic microangiopathy. Sem Hematol 1997; 34: 117-25.

13. Thompson CE, Damon LE, Ries CA, Linker CA. Thrombotic microangio-pathies in the 1980's: Clinical features, response to treatment, and the im-pact of the human immunodeficiency virus epidemic. Blood 1992; 80:1890-95.

14. Ucar A, Fernandez HF, Byrnes JJ, Lian ECY, Harrington WJ. Thromboticmicroangiopathy and retroviral infections: A 13 year experience. Am J Hematol 1994; 45: 304-9.

15. Walsh C, Kriegel R, Lennette E, Karpatkin S. Thrombocytopenia in homosexual patients. Prognosis, response to therapy, and prevalence ofantibody to the retrovirus associated with the acquired immunodeficiencysyndrome. Ann Intern Med 1985; 103: 542-5.

16. Murphy MF, Metcalfe P, Waters AH. Incidence and mechanism of neu-tropenia and thrombocytopenia in patients with human immunodeficiencyvirus infection. Brit J Haematol 1987; 66: 337-40.

17. Mientjes CHC, Van Ameijden EJC, Mulder JW, Van den Hoek JAR, Continho RA, Von dem Borne AE. Prevalence of thrombocytopenia inHIV-infected and non-HIV-infected drug abusers and homosexuals. Br J Haematol 1992; 82: 615-9.

18. Landonio G, Gall M, Nosari A, et al. HIV-related severe thrombocyto-penia in intravenous drug abusers: Prevalence, response to therapy in a medium term follow-up and pathogenetic evaluation. AIDS 1998; 4: 29-34.

19. Auch D, Budde U, Hammerstein U, et al. FcR-mediated clearance inthrombopenic and non-thrombopenic patients with hemophilia A and possible relation of thrombopenia to HIV seropositivity. Eur J Haematol1987; 39: 440-6.

20. Ehmann WC, Rabkin CS, Eyster ME, Goedert JJ. Thrombocytopenia inHIV-infected and uninfected hemophiliacs. Am J Hematol 1997; 54: 296-300.

21. Holzman RS, Walsh CM, Karpatkin S. Risk for the Acquired Immunode-ficiency Syndrome among thrombocytopenic and non-thrombocytopenichomosexuals non-seropositive for Human Immunodeficiency Virus. AnnIntern Med 1987; 106: 383-6.

22. Abrams DI, Kiprov DD, Goedert JJ, et al. Antibodies to human T lym-photropic virus type III and development of the acquired immunodeficien-cy syndrome in homosexual men presenting with immune thrombocyto-penia. Ann Intern Med 1986; 105: 47-50.

23. Goldsweig HG, Grossman R, William D. Thrombocytopenia in homo-sexual men. Am J Hematol 1986; 21: 243-7.

24. Hymes KB, Greene JB, Karpatkin S. The effect of azidothymidine onHIV-related thrombocytopenia. N Engl J Med 1988; 318: 516-7.

25. Swiss Group for Clinical Studies on AIDS. Zidovudine for the treatmentof thrombocytopenia associated with HIV. A prospective study. Ann Intern Med 1988; 109: 718.

26. Arranz Caso JA, Sanchez Mingo C, Garcia Tena J. Effect of highly activeantiretroviral therapy on thrombocytopenia in patients with HIV infection[letter]. N Engl J Med 1999; 341: 1239-40.

27. Bussel J, Haimi J. Isolated thrombocytopenia in patients infected withHIV: Treament with intravenous gammaglobulin. Am J Hematol 1988;28: 79-84.

28. Oksenhendler E, Bierling P, Farcet J-P, et al. Response to therapy in 37 pa-tients with HIV-related thrombocytopenic purpura. Br J Haematol 1987;66: 491-5.

29. Pollak AN, Janinis J, Green D. Successful intravenous immunoglobulintherapy for human immunodeficiency virus-asociated thrombocytopenia.Ann Intern Med 1988; 148: 695-7.

30. Rarick MU, Montgomery T, Groshen S, Sullivan-Halley J, Jamin D, Mazumder A, et al. Intravenous immunoglobulin in treatment of humanimmunodeficiency virus-related thrombocytopenia. Am J Hematol 1991;38: 261-6.

31. Scaradavou A, Woo B, Woloski BM, Cunningham-Rundles S, EttingerLJ, Aledort LM, et al. Intravenous anti-D treatment of immune throm-bocytopenic purpura: experience in 272 patients. Blood 1997; 89: 2689-700.

32. Karpatkin S, Nardi MA. On the mechanism of thrombocytopenia in hemophiliacs multiply transfused with AHF concentrates. J Lab Clin Med1988; 111: 441-8.

33. Karpatkin S, Nardi MA. Autoimmune anti-HIV-1gp120 antibody with anti-idiotype-like activity in sera and immune complexes of HIV-1 relatedimmunologic thrombocytopenia (HIV-1-ITP). J Clin Invest 1992; 89: 356-64.

34. Karpatkin S, Nardi MA, Hymes KB. Sequestration of anti-platelet GPIIIaantibody in Rheumatoid Factor-immune complexes of human immunode-ficiency virus 1 thrombocytopenic patients. Proceedings of the NationalAcademy of Sciences USA 1995; 92: 2263-7.

35. Karpatkin S, Nardi MA, Lennette ET, Byrne B, Poiesz B. Anti-human im-munodeficiency virus type 1 antibody complexes on platelets of seroposi-tive thrombocytopenic homosexuals and narcotic addicts. Proc Natl AcadSci USA 1988; 85: 9763-7.

36. Nardi M , Karpatkin S. Antiidiotype antibody against platelet anti-GPIIIacontributes to the regulation of thrombocytopenia in HIV-1-ITP patients.J Exper Med 2000; 191: 2093-100.

37. Nardi MA, Liu L-X, Karpatkin S. GPIIIa (49-66) is a major pathophysio-logically-relevant antigenic determinant for anti-platelet GPIIIa of HIV-1-related immunologic thrombocytopenia (HIV-1-ITP). Proceedings of theNational Academy of Sciences USA 1997; 94: 7589-94.

38. Walsh CM, Nardi MA, Karpatkin S. On the mechanism of thrombocyto-penic purpura in sexually-active homosexual men. N Engl J Med 1984;311: 635-9.

39. Yu J-R, Lennette ET, Karpatkin S. Anti-F(ab')2 antibodies in throm-bocytopenic patients at risk for Acquired Immunodeficiency Syndrome. J Clin Invest 1986; 77: 1756-61.

40. Najean Y, Rain J-D. The mechanism of thrombocytopenia in patients withHIV. J Lab Clin Med 1994; 123: 415.

41. Katsarou O, Terpos E, Patsouris E, Peristeris P, Viniou N, Kapsimali V, et al. Myelodysplastic features in patients with long-term HIV infectionand haemophilia. Haemophilia 2001; 7: 47-52.

Page 12: Platelet and Coagulation - UW Hematology Protocolswilliams.medicine.wisc.edu/hivcoag.pdf · Platelet and Coagulation Defects Associated ... and hemolytic uremic syndrome ... An analysis

400

Thromb Haemost 2002; 88: 389–401

42. Young G, Loechelt BJ, Rakusan TA, Nichol JL, Luban NL. Thrombo-poietin-levels in HIV-associated thrombocytopenia in children. J Pedia-trics 1998; 133: 765-9.

43. Bel-Ali Z, Dufour V, Najean Y. Platelet kinetics in human immuno-deficiency virus induced thrombocytopenia. Amer J Hematol 1987; 26:229-304.

44. Van Wyk V, Kotze HF, Heyns AP. Kinetics of indium-111-labelled plate-lets in HIV-infected patients with and without associated thrombocyto-paenia. Eur J Haematol 1999; 62: 332-5.

45. Siegel RS, Rae JL, Kessler CM. Immune thrombocytopenic purpura inHTLV-III men. Blood 1986; 68 (Suppl 1): 134A.

46. Ballem P, Belzberg A, Devine D, Lyster D, Spruston B, Chambers H, et al.Kinetics studies of the mechanism of thrombocytopenia in patients withhuman immunodeficiency virus infection. N Engl J Med 1992; 327: 1779-84.

47. Cole JL, Marzec UM, Gunthel CJ, Karpatkin S, Worford L, Sundell IB, et al. Ineffective platelet production in thrombocytopenic human immuno-deficiency virus-infected patients. Blood 1998; 91: 3239-46.

48. Stella CC, Ganser A, Hoezler D. Defective in vitro growth of the hemo-poietic progenitor cells in the acquired immunodeficiency syndrome. J Clin Invest 1987; 80: 286.

49. Chelucci C, Federico M, Guerriero R, G. M, Casella I, Pelosi E, et al. Pro-ductive human immunodeficiency virus-1 infection of purified mega-karyocytic progenitors/precursors and maturing megakaryocytes. Blood1998; 91: 1225-34.

50. Zucker-Franklin D, Cao Y. Megakaryocytes of human immunodeficiencyvirus-infected individuals express viral RNA. Proc Natl Acad Sci USA1989; 86: 5595.

51. Basch R, Kouri YH, Karpatkin S. Expression of CD4 by human mega-karyocytes. Proc Natl Acad Sci 1990; 87: 8085-9.

52. Kouri YH, Borkowsky W, Nardi M, Karpatkin S, Basch R. Human mega-karyocytes have a CD4 molecule capable of binding human immuno-deficiency virus-1. Blood 1993; 81: 2664-70.

53. Kowalska MA, Ratajczak J, Hoxie J, Brass LF, Gewirtz A, Poncz M, et al.Megakaryocyte precursors, megakaryocytes and platelets express the HIVco-receptor CXCR4 on their surface: determination of response to stro-mal-derived factor-1 by megakaryocytes and platelets. Br J Haematol1999; 104: 220-9.

54. Riviere C, Subra F, Cohen-Solal K, Cordette-Lagarde V, Letestu R,Auclair C, et al. Phenotypic and functional evidence for the expression of CXCR4 receptor during megakaryocytopoiesis. Blood 1999; 93: 1511-23.

55. Sakaguchi M, Sato T, Groopman J. Human immunodeficiency virus infection of megakaryocytic cells. Blood 1991; 77: 481.

56. Bahner I, Kearns K, Couthino S, Leonard EH, Kohn DB. Infection of human marrow stroma by HIV-1 is both required and sufficient for HIV-1induced hematopoietic suppression in vitro: demonstration by gene modi-fication of primary human stroma. Blood 1997; 90: 1787-98.

57. Davis BR, Marx JC, Johnson CE, et al. Absent or rare HIV-1 infection ofbone marrow stem/progenitor cells in vivo. J Virol 1991; 65: 1985.

58. Louache F, Henri A, Bettaieb A, et al. Role of human immunodeficiencyvirus replication in defective in vitro growth of hematopoietic progenitors.Blood 1992; 80: 2991.

59. Moses A, Nelson J, Bagby GC, Jr. The influence of human immuno-deficiency virus 1 on hematopoiesis. Blood 1998; 91: 1479-91.

60. Moses AV, Williams S, Heneveld ML, et al. Human immunodeficiencyvirus infection of bone marow endothelium reduces induction of stromalhematopoietic growth factors. Blood 1996; 87: 919-25.

61. Scadden DT, Zeira M, Woon A, et al. Human HIV-1 infection of humanbone marrow stromal fibroblasts. Blood 1990; 76: 317.

62. Steinberg HN, Anderson J, Crumpacker CS, Chatis PA. HIV infection ofthe BS-1 human stromal cell line: effect on murine hematopoiesis. Viro-logy 1993; 193: 524-7.

63. von Laer D, Hufert FT, Fenner TE, et al. CD34+ hematopoietic progenitorcells are not a major reservoir of the HIV-1 virus. Blood 1990; 76: 1281.

64. Von Laer D, Hufert FT, Fenner TE, et al. Progenitor cells are not a majorreservoir of the human immunodeficiency virus. Blood 1990; 76: 1281-6.

65. Zauli G, Carla-Re M, Davis B, et al. Impaired in vitro growth of purified(CD34+) hematopoietic progenitors in human immunodeficiency virus-1seropositive thrombocytopenic individuals. Blood 1992; 79: 2680.

66. Zauli G, Catani L, Gibellini D, et al: Impaired survival of bone marrowGPIIb/GPIIIa+ megakaryocytic cells as an additional pathogenesis mecha-nism of HIV-1-related thrombocytopenia. Br J Haematol 1996; 92: 711-7.

67. Zucker-Franklin D, Termin CS, Cooper MC. Structural change in the megakaryocytes of patients infected with human immune deficiency virus(HIV-1). Am J Pathol 1989; 134: 1295.

68. Carlo-Stella C, Ganser A, Hoelzer D. Defective in vitro growth of the hematopoietic progenitor cells in the acquired immunodeficiency syndro-me. J Clin Invest 1987; 80: 286.

69. Ishibashi T, Miller SL, Burstein SA. Type � transforming growth factor isa potent direct inhibitor of murine megakaryocytopoiesis in vitro. Blood1987; 69: 1737-41.

70. Kuter DJ, Gminski DM, Rosenberg RD. Transforming growth factor b inhibits megakaryocyte growth and endomitosis. Blood 1992; 9: 619-26.

71. Mitjavila MT, Vinci G, Villeval JL, et al. Human platelet alpha granulescontain a non-specific inhibitor of megakaryocyte colony formation: its relationship to type b transforming growth factor (TGF-b). J Cell Physiol1988; 134: 93-100.

72. Zauli G, Davis B, Re MC, et al. tat protein stimulates production of TGFb-1 by marrow macrophages: a potential mechanism for human HIV-1-indu-ced hematopoietic suppression. Blood 1992; 80: 3036.

73. Gewirtz AM, Calabretta B, Rucinski B, Niewiarowski S, Wen Y-X. Inhi-bition of human megakaryocytopoiesis in vitro by platelet factor 4 (PF4)and a synthetic COOH-terminal PF4 peptide. J Clin Invest 1989; 83: 1477.

74. Guarini A, Sanavio F, Novarino A, et al. Thrombocytopenia in acute leu-kemia patients treated with IL-2: cytolytic effect of LAK on mega-karyocytic progenitors. Br J Haematol 1991; 79: 451.

75. Volkers B, Ganser A, Greher J, et al. Effect of tumor necrosis factor on hu-man hematopoietic precursor cells. Onkolgie 1989; 12: 109.

76. Ganser A, Carlo-Stella C, Greher J, et al. Effect of recombinant interferonsalpha and gamma on human bone marrow-derived megakaryocytic proge-nitor cells. Blood 1987; 70: 1171.

77. Sonoda Y, Kuzuyama Y, Tanaka S, et al. Human IL-4 inhibits proliferati-on of megakaryocyte progenitor cells in culture. Blood 1993; 81: 624.

78. Levy GG, Nichols WC, Lian EC, Foroud T, McClintick JN, et al. Mutati-ons in a member of the ADAMTs gene family cause thrombotic throm-bocytopenic purpura. Nature 2001; 413: 488-94.

79. Furlan M, Rodolfo R, Max S, Lammle B. Acquired deficiency of von Wil-lebrand factor-cleaving protease in a patient with thrombotic throm-bocytopenic purpura. Blood 1998; 91: 2839-46.

80. Tsai HM, Lian EC. Antibodies to von Willebrand factor-cleaving proteasein acute thrombotic thrombocytoenic purpura. N Engl J Med 998; 339:1584-94.

81. Tsai HM, Chandler WL, Sarode R, Hoffman R, Jelacic S, Habeeb RL, et al. von Willebrand factor and von Willebrand factor-cleaving metallo-protease activity in Escherichia coli 0157:H7-associated hemolytic uremicsyndrome. Pediatric Res 2001; 49: 653-9.

82. Arai S, Allan C, Streiff M, Hutchins GM, Vogelsang GB, Tsai HM. VonWillebrand factor-cleaving protease activity and proteolysis of von Wille-brand factor in bone marrow transplant-associated thrombotic microangio-pathy. Hematol J 2001; 2: 292-9.

83. Sahud MA, Claster S, Liu L, Ero M, Harris K, Furlan M. von Willebrandfactor-cleaving protease inhibitor in a patient with human immuno-deficiency syndrome-associated thrombotic thrombocytopenic purpura. B J Haematol 2002; 116: 909-11.

84. Van Der Lelie J, Lange JMA, J.J.E. V, et al. Autoimmunity against bloodcells in human immunodeficiency virus (HIV) infection. Br J Haematol1987; 67: 109.

85. Gonzalez-Conejero R, Rivera J, Rosillo MC, Cano A, Rodriguez Y, Vin-cente V. Association of autoantibodies against platelet glycoproteins Ib/IX

Page 13: Platelet and Coagulation - UW Hematology Protocolswilliams.medicine.wisc.edu/hivcoag.pdf · Platelet and Coagulation Defects Associated ... and hemolytic uremic syndrome ... An analysis

401

Karpatkin et al.: Platelet and Coagulation Defects Associated with HIV-1-Infection

and IIb/IIIa and platelet-reactive anti-HIV antibodies in thrombocytopenicnarcotic addicts. Br J Haematol 1996; 93: 464-71.

86. Karpatkin S, Nardi MA, Kouri Y. Internal image anti-idiotype HIV-1gp120 antibody in human immunodeficiency virus 1 (HIV-1)-seropositi-ve individuals with thrombocytopenia. Proc Natl Acad Sci USA 1992; 89:1487-91.

87. Bettaieb A, Oksenhendler E, Duedari N, Bierling P. Cross-reactive antibo-dies between HIV-gp120 and platelet gpIIIa (CD61) in HIV-related im-mune thrombocytopenic purpura. Clin Exp Immunol 1996; 103: 19-23.

88. Chia WK, Blanchette V, Mody M, Wright JF, Freedman J: Characterizati-on of HIV-1-specific antibodies and HIV-1-crossreactive antibodies toplatelets in HIV-1-infected haemophiliac patients. Br J Haematol 1998;103: 1014-22.

89. Kouri Y, Basch RS, Karpatkin S. B-cell subsets and platelet counts inHIV-1 seropositive subjects. Lancet 1992; 339: 1445-6.

90. Burastero SE, Casali P. Characterization of human CD5 (Leu-1, OKT1)+B lymphocytes and the antibodies they produce in B lymphocytes: func-tion and regulation. Microbiol Immunol 1989; 11: 231-62.

91. Feffer SE, Fox FL, Orsen MM, et al. Thrombotic tendencies and correlati-on with clinical status in patients infected with HIV. South Med J 1995;88: 1126-30.

92. Stahl CP, Widerman CS, Spira TJ, et al. Protein S deficiency in men withlong-term human immunodeficiency virus infection. Blood 1993; 81:1801-7.

93. Toulon P, Lamine M, Ledjev I, Guez T. Heparin cofactor II deficiency inpatients infected with the human immunodeficiency virus. Thromb Hae-most 1993; 70: 730-5.

94. Bissuel F, Berruyer M, Causse, et al. Acquired protein S deficiency: Correlation with advanced disease in HIV-1-infected patients. J AcquirImmune Defic Syndr 1992; 5: 484-9.

95. Lafeuillade A, Alssi MC, Poizot-Martin I, et al. Protein S deficiency andHIV infection. N Engl J Med 1991; 1220.

96. Hassell KL, Dressin DC, Neuman A, Ellsion R, Marlar RA. Correlation ofantiphospholipid antibodies and protein S deficiency with thrombosis inHIV-1infected men. Blood Coagul Fibrinolysis 1994; 5: 455-62.

97. Janier M, Flageul B, Drouet L, Scobohaci ML, Villette JM, Palangie A, et al. Cutaneous and plasma levels of von Willebrand factor in AIDS: a marker of endothelial stimulation. J Invest Dermatol 1988; 90: 703-7.

98. Scadden DT, Zon LI, Groopman JE. Pathophysiology and management ofHIV-associated hematologic disorders. Blood 1989; 74: 1455-63.

99. Canoso RT, Zon LI, Groopman JE. Anticardiolipin antibodies associatedwith HTLV-III infection. Br J Haematol 1987; 65: 494-8.

100. Bloom EJ, Abrams DI, Rodgers GM. Lupus anticoagulant in the acquiredimmunodeficiency syndrome. JAMA 1986; 256: 491-3.

101. Cohen AJ, Philips TM, Kessler CM. Circulating coagulation inhibitors inthe acquired immunodeficiency syndrome. Ann Intern Med 1986; 104:175-80.

102. Leder AN, Flansbaum B, Zandman-Goddard G, Asherson R, Shoenfeld Y.Antiphospholipid syndrome induced by HIV. Lupus 2001; 10: 370-4.

103. Abuaf N, Laperche S, et al. Autoantibodies to phospholipids and to the coagulation proteins in AIDS. Thromb Haemost 1997; 77: 856-61.

104. Shapiro SS, Thiagarajan P. Lupus anticoagulants. Prog Hemost Thromb1982; 6: 263-85.

105. Bajaj SP, Rapaport SI, Barclay S, Herbst KD. Acquired hypoprothrombi-nemia due to non-neutralizing antibodies to prothrombin: mechanisms andmanagement. Blood 1985; 65: 1538-43.

106. Cohen JR, Lackner R, Wenig P, et al. Deep venous thrombosis in patientswith AIDS. NY State J Med 1991; 90: 159-61.

107. Saber AA, Aboolian A, LaRaja RD, Baron H, Hanna K. HIV/AIDS andthe risk of deep vein thrombosis: A study of 45 patients with lower extre-mity involvement. Am Surg 2001; 67: 645-7.

108. Saif MW, Bona R, Greenberg B. AIDS and thrombosis: Retrospective study of 131 HIV-infected patients. AIDS Patient Care STDS 2001; 15:311-20.

109. Friedman SM, Margo CE. Bilateral retinal central vein occlusions in a patient with acquired immunodeficiency syndrome. Arch Ophthalmol1988; 106: 1508-9.

110. Doberson MJ, Kleinschmidt-DeMasters BK. Superior sagittal sinusthrombosis in a patient with acquired immunodeficiency syndrome. ArchPath Lab Med 1994; 118: 844-6.

111. Lee CA. The natural history of HIV disease in haemophilia. Blood Rev1998; 12: 135-44.

112. Bray PF, Kroner BL, Arkin S, et al. Loss of high responder inhibitors inpatients with severe hemophilia A and human immunodeficiency virus type 1 infection: A report from the multi-center hemophilia cohort study.Am J Hematol 1993; 42: 375.

113. Wilde JT. Protease inhibitor therapy and bleeding. Haemophilia 2000; 6:487-90.

114. Racoosin JA, Kessler CM. Bleeding episodes in HIV-positive hemophi-liacs taking protease inhibitors: A case series. Hemophilia 1999; 5: 266-9.

115. Pollman H, Richter H, Jurgens H. Platelet dysfunction as the cause ofspontaneous bleeding in two hemophiliac patients taking protease inhibi-tors. Thromb Haemost 1998; 79: 1213-4.

116. Nielsen H. Hypermenorrhea associated with ritonavir. Lancet 1999; 353:811-2.

Received May 2, 2002 Accepted June 17, 2002