Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Haematologica ; 105(7): 1791-1801, 2020 07.
Article in English | MEDLINE | ID: mdl-32381574

ABSTRACT

Acquired hemophilia A (AHA), a rare bleeding disorder caused by neutralizing autoantibodies against coagulation factor VIII (FVIII), occurs in both men and women without a previous history of bleeding. Patients typically present with an isolated prolonged activated partial thromboplastin time due to FVIII deficiency. Neutralizing antibodies (inhibitors) are detected using the Nijmegen-modified Bethesda assay. Approximately 10% of patients do not present with bleeding and, therefore, a prolonged activated partial thromboplastin time should never be ignored prior to invasive procedures. Control of acute bleeding and prevention of injuries that may provoke bleeding are top priorities in patients with AHA. We recommend treatment with bypassing agents, including recombinant activated factor VII, activated prothrombin complex concentrate, or recombinant porcine FVIII in bleeding patients. Autoantibody eradication can be achieved with immunosuppressive therapy, including corticosteroids, cyclophosphamide and rituximab, or combinations thereof. The median time to remission is 5 weeks, with considerable interindividual variation. FVIII activity at presentation, inhibitor titer and autoantibody isotype are prognostic markers for remission and survival. Comparative clinical studies to support treatment recommendations for AHA do not exist; therefore, we provide practical consensus guidance based on recent registry findings and the authors' clinical experience in treating patients with AHA.


Subject(s)
Hemophilia A , Animals , Autoantibodies , Blood Coagulation Tests , Factor VIII , Female , Hemophilia A/diagnosis , Hemophilia A/drug therapy , Hemorrhage , Humans , Male , Rituximab/therapeutic use , Swine
2.
J Thromb Haemost ; 18(1): 36-43, 2020 01.
Article in English | MEDLINE | ID: mdl-31448877

ABSTRACT

BACKGROUND: Recombinant porcine factor VIII (rpFVIII, OBI-1, susoctocog alfa) is used for the treatment of acute bleeds in patients with acquired hemophilia A (AHA). Inhibitors in AHA can sometimes cross-react with rpFVIII. OBJECTIVES: To assess the frequency, strength, and determinants of cross-reactivity. PATIENTS/METHODS: Baseline samples from 70 patients of the prospective, observational cohort study GTH-AH 01/2010 were assessed for anti-human FVIII and anti-rpFVIII inhibitors using modified Nijmegen-Bethesda assays, as well as anti-human FVIII domain reactivity using enzyme-linked immunoassay (ELISA). RESULTS: Anti-human FVIII inhibitors were present in all samples ranging between 0.7 and 3891 Bethesda Units (BU)/mL. Inhibitors from 31 of 70 patients (44%) partially inhibited rpFVIII with anti-rpFVIII titers ranging between 0.5 and 471 BU/mL. Anti-rpFVIII titers were ≤5 BU in most patients. Patients with cross-reacting inhibitors, as compared to patients without, had significantly higher anti-human FVIII titers (27.8 versus 5.4 BU/mL) and lower baseline FVIII activity (<1 versus 2.6 IU/dL). The ratio between anti-rpFVIII to anti-human titers was highest for inhibitors involving the C1 domain. Cross-reactivity was very rare, if inhibitors reacted only with the C2 domain of FVIII (6%). An anti-human FVIII titer of >100 BU/mL predicted cross-reactivity with 97% likelihood, whereas an anti-human FVIII titer of <3.8 BU/mL predicted absent cross-reactivity with 90% likelihood. CONCLUSION: Cross-reacting inhibitors should be considered when choosing a treatment for bleeding patients with AHA. Cross-reactivity is frequent in patients with anti-human FVIII titers of >100 BU/mL.


Subject(s)
Factor VIII , Hemophilia A , Animals , Blood Coagulation Tests , Cross Reactions , Hemophilia A/diagnosis , Hemophilia A/drug therapy , Humans , Prospective Studies , Swine
3.
Blood ; 125(7): 1091-7, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25525118

ABSTRACT

Acquired hemophilia A (AHA) is caused by autoantibodies against factor VIII (FVIII). Immunosuppressive treatment (IST) results in remission of disease in 60% to 80% of patients over a period of days to months. IST is associated with frequent adverse events, including infections as a leading cause of death. Predictors of time to remission could help guide IST intensity but have not been established. We analyzed prognostic factors in 102 prospectively enrolled patients treated with a uniform IST protocol. Partial remission (PR; defined as no active bleeding, FVIII restored >50 IU/dL, hemostatic treatment stopped >24 hours) was achieved by 83% of patients after a median of 31 days (range 7-362). Patients with baseline FVIII <1 IU/dL achieved PR less often and later (77%, 43 days) than patients with ≥1 IU/dL (89%, 24 days). After adjustment for other baseline characteristics, low FVIII remained associated with a lower rate of PR (hazard ratio 0.52, 95% confidence interval 0.33-0.81, P < .01). In contrast, PR achieved on steroids alone within ≤21 days was more common in patients with FVIII ≥1 IU/dL and inhibitor concentration <20 BU/mL (odds ratio 11.2, P < .0001). Low FVIII was also associated with a lower rate of complete remission and decreased survival. In conclusion, presenting FVIII and inhibitor concentration are potentially useful to tailor IST in AHA.


Subject(s)
Hemophilia A , Immunosuppressive Agents/therapeutic use , Adult , Aged , Aged, 80 and over , Autoantibodies/adverse effects , Factor VIII/analysis , Factor VIII/immunology , Female , Hemophilia A/diagnosis , Hemophilia A/mortality , Hemophilia A/therapy , Humans , Male , Middle Aged , Prognosis , Remission Induction , Survival Analysis , Treatment Outcome
4.
Thromb Res ; 134 Suppl 1: S48-52, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24968961

ABSTRACT

Seventy years ago, the average life expectancy for patients with severe haemophilia A was less than 17 years. Today, due to the availability of safe and effective clotting factor concentrates, life expectancy is nearly normal, at least in patients without viral infections. More individuals are living into their 70s and 80s, acquiring a range of diseases that are common in elderly persons. One of the most important challenges includes the treatment of comorbidities, especially cardiovascular diseases. Although most evidence suggests that haemophilia, at least the severe manifestation, partially protects against myocardial infarction, stroke and venous thromboembolism, typical cardiovascular risk factors can still be present despite the clotting defect. Patients with haemophilia are equally or even more prone to obesity, hypertension, diabetes, and dyslipidaemia, and this is especially true for HIV-infected individuals using highly active antiretroviral therapy. The management of elderly haemophilia patients with cardiovascular comorbidities is hampered by a lack of evidence-based guidelines. Nevertheless, experience in treating cardiovascular disease is growing amongst the haemophilia community, and several authors have published their own recommendations for managing a variety of commonly encountered cardiovascular scenarios in haemophilia patients. Basic recommendations exist for risk-factor management, the adaptation of factor replacement therapy in the at-risk elderly, management of coronary revascularization, the management of acute coronary syndrome and atrial fibrillation. This review outlines our current knowledge about cardiovascular risk in elderly haemophilia patients, recommendations for clinical decision making, and our own experiences of managing individuals with coronary heart disease and atrial fibrillation.


Subject(s)
Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/therapy , Hemophilia A/epidemiology , Hemophilia A/therapy , Age Factors , Comorbidity , Hemophilia A/complications , Humans , Risk Factors
5.
Biores Open Access ; 2(6): 399-411, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24380050

ABSTRACT

Hemophilia patients infected with human immunodeficiency virus (HIV) 30 years ago show increased proportions of activated CD8(+)DR(+) blood lymphocytes. We hypothesized that this might indicate a cellular immune response directed against HIV and might be the reason for long-term clinical stability of these patients. CD8(+) peripheral blood lymphocytes (PBL) reactive with six HIV and two cytomegalovirus (CMV) pentamers were determined in heparinized whole blood. Additional lymphocyte subsets as well as plasma cytokines and HIV-1 load were studied. Long-term HIV-infected hemophilia patients with (n=15) or without (n=33) currently detectable HIV-1 load in the plasma showed higher proportions of CD8(+) lymphocytes reactive with HIV (p<0.001) and CMV pentamers (p=0.010) than healthy individuals. The cellular anti-HIV response tended to be stronger and more polyclonal in patients during periods of viral replication than in patients with retroviral quiescence (p=0.077). Anti-HIV CD8(+) lymphocyte responses were strongest in patients with high counts of activated CD8(+)DR(+) T (r=0.353; p=0.014) and low CD19(+) B lymphocyte counts (r=-0.472; p=0.001). Patients with or without HIV-1 viral load showed normal Th1 and Th2 plasma cytokine levels and high plasma interleukin-6 (versus healthy controls, p=0.001) and tumor necrosis factor-α (p=0.020). Hemophilia patients who have been living with HIV for more than 30 years showed a polyclonal CD8(+) T-cell response against HIV and CMV. This cellular antiviral immune response was strongest during periods of HIV-1 replication and remained detectable during periods of HIV-1 quiescence. We hypothesize that the consistent cellular anti-HIV-1 response in combination with highly active antiretroviral therapy ensures stability and survival of these chronically HIV-1-infected hemophilia patients.

6.
Blood ; 120(1): 39-46, 2012 Jul 05.
Article in English | MEDLINE | ID: mdl-22618709

ABSTRACT

Acquired hemophilia A is a rare bleeding disorder caused by autoantibodies to coagulation FVIII. Bleeding episodes at presentation are spontaneous and severe in most cases. Optimal hemostatic therapy is controversial, and available data are from observational and retrospective studies only. The EACH2 registry, a multicenter, pan-European, Web-based database, reports current patient management. The aim was to assess the control of first bleeding episodes treated with a bypassing agent (rFVIIa or aPCC), FVIII, or DDAVP among 501 registered patients. Of 482 patients with one or more bleeding episodes, 144 (30%) received no treatment for bleeding; 31 were treated with symptomatic therapy only. Among 307 patients treated with a first-line hemostatic agent, 174 (56.7%) received rFVIIa, 63 (20.5%) aPCC, 56 (18.2%) FVIII, and 14 (4.6%) DDAVP. Bleeding was controlled in 269 of 338 (79.6%) patients treated with a first-line hemostatic agent or ancillary therapy alone. Propensity score matching was applied to allow unbiased comparison between treatment groups. Bleeding control was significantly higher in patients treated with bypassing agents versus FVIII/DDAVP (93.3% vs 68.3%; P = .003). Bleeding control was similar between rFVIIa and aPCC (93.0%; P = 1). Thrombotic events were reported in 3.6% of treated patients with a similar incidence between rFVIIa (2.9%) and aPCC (4.8%).


Subject(s)
Hemophilia A/drug therapy , Hemorrhage/drug therapy , Hemostatics/therapeutic use , Registries/statistics & numerical data , Adolescent , Adult , Aged , Aged, 80 and over , Blood Coagulation Factors/therapeutic use , Deamino Arginine Vasopressin/therapeutic use , Europe/epidemiology , Factor VIII/therapeutic use , Factor VIIa/therapeutic use , Female , Hemophilia A/mortality , Hemorrhage/mortality , Humans , Incidence , Male , Middle Aged , Recombinant Proteins/therapeutic use , Treatment Outcome , Young Adult
7.
Blood ; 120(1): 47-55, 2012 Jul 05.
Article in English | MEDLINE | ID: mdl-22517903

ABSTRACT

Acquired hemophilia A (AHA) is an autoimmune disease caused by an autoantibody to factor VIII. Patients are at risk of severe and fatal hemorrhage until the inhibitor is eradicated, and guidelines recommend immunosuppression as soon as the diagnosis has been made. The optimal immunosuppressive regimen is unclear; therefore, data from 331 patients entered into the prospective EACH2 registry were analyzed. Steroids combined with cyclophosphamide resulted in more stable complete remission (70%), defined as inhibitor undetectable, factor VIII more than 70 IU/dL and immunosuppression stopped, than steroids alone (48%) or rituximab-based regimens (59%). Propensity score-matched analysis controlling for age, sex, factor VIII level, inhibitor titer, and underlying etiology confirmed that stable remission was more likely with steroids and cyclophosphamide than steroids alone (odds ratio = 3.25; 95% CI, 1.51-6.96; P < .003). The median time to complete remission was approximately 5 weeks for steroids with or without cyclophosphamide; rituximab-based regimens required approximately twice as long. Immunoglobulin administration did not improve outcome. Second-line therapy was successful in approximately 60% of cases that failed first-line therapy. Outcome was not affected by the choice of first-line therapy. The likelihood of achieving stable remission was not affected by underlying etiology but was influenced by the presenting inhibitor titer and FVIII level.


Subject(s)
Autoantibodies/immunology , Factor VIII/immunology , Hemophilia A/drug therapy , Immunosuppressive Agents/administration & dosage , Registries/statistics & numerical data , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antibodies, Monoclonal, Murine-Derived/adverse effects , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Cyclosporine/administration & dosage , Cyclosporine/adverse effects , Europe , Factor VIII/therapeutic use , Female , Follow-Up Studies , Humans , Immunosuppressive Agents/adverse effects , Male , Middle Aged , Rituximab , Secondary Prevention , Steroids/administration & dosage , Steroids/adverse effects , Treatment Outcome
8.
BMC Res Notes ; 3: 161, 2010 Jun 07.
Article in English | MEDLINE | ID: mdl-20529258

ABSTRACT

BACKGROUND: Acquired hemophilia A (AHA) is a rare bleeding disorder caused by an autoantibody to coagulation factor (F) VIII. It is characterized by soft tissue bleeding in patients without a personal or family history of bleeding. Bleeding is variable, ranging from acute, life-threatening hemorrhage, with 9-22% mortality, to mild bleeding that requires no treatment. AHA usually presents to clinicians without prior experience of the disease, therefore diagnosis is frequently delayed and bleeds under treated. METHODS: Structured literature searches were used to support expert opinion in the development of recommendations for the management of patients with AHA. RESULTS: Immediate consultation with a hemophilia center experienced in the management of inhibitors is essential to ensure accurate diagnosis and appropriate treatment. The laboratory finding of prolonged activated partial thromboplastin time with normal prothrombin time is typical of AHA, and the diagnosis should be considered even in the absence of bleeding. The FVIII level and autoantibody titer are not reliable predictors of bleeding risk or response to treatment. Most patients with AHA are elderly; comorbidities and underlying conditions found in 50% of patients often influence the clinical picture. Initial treatment involves the control of acute bleeding with bypassing agents. Immunosuppressive treatment to eradicate the FVIII inhibitor should be started as soon as the diagnosis is confirmed to reduce the time the patient is at risk of bleeding. CONCLUSIONS: These recommendations aim to increase awareness of this disorder among clinicians in a wide range of specialties and provide practical advice on diagnosis and treatment.

9.
Viral Immunol ; 23(1): 87-97, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20121406

ABSTRACT

After initiation of highly-active antiretroviral therapy (HAART), long-term HIV-infected hemophilia patients have been shown to lose autoantibodies against CD4(+) peripheral blood leukocytes (PBL), suggesting that HAART induces autoimmunity-blocking mechanisms. We compared cytokine levels and subpopulations of lymphocytes and dendritic cells (DC) in the blood of 40 long-term HIV(+) patients with those of 13 long-term HIV(-) hemophilia patients; 23 HIV(+) patients had a detectable retroviral load. Cell subsets were determined using flow cytometry and cytokine levels were measured using ELISA. HIV(+) patients showed higher proportions of DC subpopulations with immunostimulatory phenotypes (p < 0.01), CD8(+) PBL (p < 0.001), and IL-2 (p < 0.001) and sIL-2R plasma levels (p = 0.002) than HIV(-) patients. They also exhibited increased proportions of T PBL with immunosuppressive phenotypes such as CD3(+)CD4(+)CD25(+)Foxp3(+) (p = 0.001), and CD3(+)CD8(+)CD28(-)Foxp3(+) PBL (p < 0.001), and a decreased IL-7R expression on CD3(+)CD8(+) PBL (p = 0.001) compared to HIV(-) patients. Frequencies of CD3(+)CD4(+)CD25(+) PBL producing IL-2, IL-4, IL-10, IL-12, and/or IFN-gamma, and of CD3(+)CD4(+)CD28(-) PBL secreting IL-2 and/or IL-4 were lower in HIV(+) than in HIV(-) patients (p

Subject(s)
Antiretroviral Therapy, Highly Active , Autoantibodies/blood , Dendritic Cells/immunology , HIV Infections/immunology , HIV Long-Term Survivors , Hemophilia A/complications , T-Lymphocyte Subsets/immunology , Adult , Antigens, CD/analysis , Cytokines/biosynthesis , Dendritic Cells/chemistry , Forkhead Transcription Factors/analysis , HIV Infections/complications , HIV Infections/drug therapy , Humans , Male , Middle Aged , T-Lymphocyte Subsets/chemistry , Young Adult
11.
Haematologica ; 94(4): 566-75, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19336751

ABSTRACT

Acquired hemophilia A (AHA) is a rare bleeding disorder characterized by autoantibodies directed against circulating coagulation factor (F) VIII. Typically, patients with no prior history of a bleeding disorder present with spontaneous bleeding and an isolated prolonged aPTT. AHA may, however, present without any bleeding symptoms, therefore an isolated prolonged aPTT should always be investigated further irrespective of the clinical findings. Control of acute bleeding is the first priority, and we recommend first-line therapy with bypassing agents such as recombinant activated FVII or activated prothrombin complex concentrate. Once the diagnosis has been achieved, immediate autoantibody eradication to reduce subsequent bleeding risk should be performed. We recommend initial treatment with corticosteroids or combination therapy with corticosteroids and cyclophosphamide and suggest second-line therapy with rituximab if first-line therapy fails or is contraindicated. In contrast to congenital hemophilia, no comparative studies exist to support treatment recommendations for patients with AHA, therefore treatment guidance must rely on the expertise and clinical experience of specialists in the field. The aim of this document is to provide a set of international practice guidelines based on our collective clinical experience in treating patients with AHA and contribute to improved care for this patient group.


Subject(s)
Hemophilia A/diagnosis , Hemophilia A/drug therapy , Drug Therapy, Combination , Germany , Hemophilia A/etiology , Hemorrhage/drug therapy , Hemorrhage/prevention & control , Humans , International Cooperation , Partial Thromboplastin Time
12.
Ann Hematol ; 88(4): 365-70, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19083235

ABSTRACT

Acquired haemophilia is an autoimmune disorder characterised by autoantibody formation against coagulation factor VIII. Immunosuppressive treatments including steroids, cytotoxic drugs, rituximab or combinations thereof have been used to eradicate autoantibodies. Very few prospective studies exist evaluating the use of these treatments. Here, we performed a survey among 73 physicians from 57 haemophilia treatment centres in order to describe current practice patterns and critical issues for future research in acquired haemophilia. The results demonstrate a high diversity of first- and second-line treatments. Factors influencing treatment decision were underlying disorder, severity of bleeding and inhibitor titre. Frequently used first-line treatments were steroids plus cyclophosphamide (44%) and steroids alone (11%). Second-line treatment was most often rituximab (30%), with or without steroids and/or cyclophosphamide. Most participants indicated to change from first- to second-line treatment after 4 weeks in case of failure to obtain partial remission (31%), continued bleeding (40%) or continued severe bleeding requiring bypass treatment (59%). Immunoadsorption was preferred for first- and second-line treatment by 10% and 9% of participants, respectively. These results highlight critical issues in the field. Open questions and directions for future research are discussed.


Subject(s)
Hemophilia A/drug therapy , Immunosuppressive Agents/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Murine-Derived , Austria , Autoantibodies/blood , Cyclophosphamide/therapeutic use , Data Collection , Germany , Hemophilia A/etiology , Humans , Immunosorbent Techniques , Practice Patterns, Physicians' , Remission Induction , Rituximab , Steroids/therapeutic use , Switzerland , Treatment Failure
13.
J Acquir Immune Defic Syndr ; 47(1): 1-15, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17971705

ABSTRACT

Defective regulatory components of the immune system seem to contribute to HIV disease progression. Highly active antiretroviral therapy (HAART) was reported to restore malfunctioning immunologic regulatory components. To corroborate this hypothesis, we studied different dendritic-cell (DC) and T-cell subsets with regulatory phenotype in 41 clinically stable patients with hemophilia more than 25 years after infection with HIV and 10 years after initiation of HAART. Compared with healthy controls, patients showed normal DC1 and DC2 levels, increased CD8 peripheral blood lymphocyte (PBL) counts and T helper (Th) 2 proportions, and decreased Th1, CD3CD4CD127, and CD3CD8CD127 PBL proportions. High viral load was associated with high DC1, whereas high CD8 PBL counts were associated with low DC2. CD3CD4CD25Foxp3 (regulatory T [Treg] cell) and CD3CD8CD28Foxp3 PBL counts (suppressor T [Ts] cell) exhibited normal levels in patients with undetectable retroviral load, were increased in parallel to retroviral load, and were associated with low CD8 T lymphocytes and low CD19 B lymphocytes. Normal or even increased levels of DCs and normal or even increased levels of PBL with a Treg or Ts phenotype that coincide with viral load increases and CD8 T- and CD19 B-lymphocyte decreases suggest a functioning immunoregulatory system that reacts to HIV replication. Increased CD8 PBL counts imply immunocompetence. Increased CD8 PBL counts and normal or even upregulated immunoregulatory cells might stabilize our long-term HIV-infected patients with hemophilia clinically.


Subject(s)
Antiretroviral Therapy, Highly Active , Dendritic Cells/immunology , HIV Infections/immunology , Hemophilia A/complications , Lymphocyte Subsets , Cytokines/blood , HIV Infections/complications , HIV Infections/drug therapy , HIV-1/genetics , HIV-1/isolation & purification , HLA-DR Antigens/immunology , Humans , Immunophenotyping , Phenotype , Receptors, Cytokine/antagonists & inhibitors , Receptors, Cytokine/blood , Viral Load
14.
J Clin Immunol ; 28(1): 58-72, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17879005

ABSTRACT

We investigated dendritic cell (DC) subsets as well as cellular and humoral immune parameters in long-term HIV-infected hemophilia patients with clinically stable disease. DC subsets were determined by their function to produce either IL-10 or IL-12. CD11c(+)CD83(+)CD40(+)IL-10(+) and CD11c(+)CD83(+)CD40(+)IL-12(+) DC were studied in freshly obtained blood samples of 28 HIV(+) and 15 HIV(-) patients and 39 healthy controls using four-color flow cytometry, and were analyzed in relation to blood lymphocyte subpopulation counts, proportions of IgG-coated CD4(+) blood lymphocytes, neopterin, and HIV-1 viral load in the plasma, and in vitro responses of patient lymphocytes to mitogens. Proportions and ratios of IL-10(+) DC and IL-12(+) DC were similar in HIV(+) and HIV(-) patients and healthy controls. Whereas IL-12(+) DC in HIV(+) patients were associated with high CD3(+)CD4(-)DR(+) lymphocyte counts, IL-10(+) DC were associated with the proportion of IgG-coated CD4(+) blood lymphocytes. These data suggest that long-term HIV-infected hemophilia patients with clinically stable disease have normal levels of functional IL-10(+) DC and IL-12(+) DC that might be involved in halting the progression of disease.


Subject(s)
Dendritic Cells/immunology , HIV Infections/immunology , HIV-1 , Hemophilia A/immunology , Interleukin-12/analysis , Lymphocyte Subsets/immunology , Adult , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Progression , Female , HIV Envelope Protein gp120/immunology , HIV Envelope Protein gp120/metabolism , HIV Infections/virology , HIV Long-Term Survivors , Hemophilia A/complications , Hemophilia A/virology , Humans , Interleukin-12/immunology , Lymphocyte Count , Male , Middle Aged , Neopterin/blood , RNA, Viral/blood
16.
AIDS Res Hum Retroviruses ; 23(7): 886-90, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17678471

ABSTRACT

It was previously reported that without highly active antiretroviral therapy (HAART), secretion of Th1 cytokines and antiviral IFN-gamma in HIV-infected patients is decreased, whereas the production of Th2 cytokines, proinflammatory cytokines, and TNF-alpha is increased. We studied the effect of HAART on Th1-, Th2-, and monocyte-derived cytokines, and on the Th2-type immune response marker soluble (s)CD30 in HIV-1-infected hemophilia patients. Viral Load (VL), CD4+ lymphocyte counts, and plasma levels of sIL-1RA, IL-2, sIL-2R, IL-3, IL-4, IL-6, sIL-6R, IL-7, IL-10, TNF-alpha, TGF-beta2, IFN-gamma, and sCD30 were measured in 18 patients who received HAART. Nine patients were initially treatment-naive and were monitored after the initiation of HAART. sCD30 median levels were significantly higher in treatment-naive patients than in patients who were on HAART (77 vs. 30 U/ml, p = 0.005). A strong association was observed between sCD30 and VL (r = 0.85, p = 0.004). After the initiation of HAART, sCD30 levels decreased and remained low (at 1 year, 38; at 2 years, 41 U/ml; p = 0.012 and p = 0.021, respectively, as compared to baseline level) and this was accompanied by a decrease in VL and monocyte-derived IL-6 and an increase in CD4+ lymphocyte counts and Th1-derived IFN-gamma. One year after the initiation of HAART a strong inverse correlation was observed between IFN-gamma and VL (r = -0.83, p = 0.006). In contrast to sCD30 and IFN-gamma, CD4 counts and plasma IL-6 did not correlate with VL at any time. Our data suggest that decreasing sCD30 and increasing IFN-gamma plasma levels are indicators of effective HAART treatment and CD4 Th1 cell recovery in HIV-infected patients.


Subject(s)
Antiretroviral Therapy, Highly Active , HIV Infections/drug therapy , Ki-1 Antigen/drug effects , Viral Load , Adult , Biomarkers , CD4-Positive T-Lymphocytes , Case-Control Studies , Cytokines/blood , HIV Infections/blood , HIV Infections/immunology , Humans , Interferon-gamma/analysis , Interferon-gamma/drug effects , Interleukin-6/metabolism , Ki-1 Antigen/blood , Male , Predictive Value of Tests , Treatment Outcome
17.
Immunol Lett ; 91(1): 23-32, 2004 Jan 30.
Article in English | MEDLINE | ID: mdl-14757366

ABSTRACT

OBJECTIVE: We have postulated that the host autoimmune response regulates and mediates CD4 depletion during HIV infection by opsonization of circulating CD4(+) lymphocytes carrying autoreactive immune complexes (IC) consisting of complement-fixing IgM and IgG, and during advanced stages of HIV disease of IgM/ IgG/gp120 complexes. In this retrospective study, we investigated whether HIV causes CD4 depletion by direct cytotoxicity or indirectly by induction of a host autoimmune response. PATIENTS AND METHODS: In 1996, 12 HIV(+) hemophilia patients were converted to highly active antiretroviral therapy (HAART), while 10 other patients were maintained on conventional antiretroviral treatment and another 11 patients refused to be treated with antiretroviral drugs. The host immune response of these 33 HIV(+) patients was studied during the periods of minimum viral replication (Interval 1), subsequent rise in viral replication with strong replication dynamic (Interval 2), and maximum viral replication (Interval 3). The patients were categorized into three groups according to viral load (VL). Group A: patients with low level VL (n=10) showed a modest increase from <80 to <4 log 10 HIV-1 RNA copies per milliliter plasma during the observation period; Group B: patients with medium level VL (n=12) showed a stronger increase from <80 to >4 log 10 copies per milliliter plasma; and Group C: patients with high level VL (n=11) consistently had a median of >4 log 10 copies per milliliter plasma, during Intervals 1-3, with the exception of one patient who during Interval 2 had 4800 copies per milliliter. Blood lymphocyte subpopulations, proportions of CD4(+) blood lymphocytes coated with IgM, IgG, C3d and/or gp120, in vitro responses to mitogens and pooled allogeneic stimulator cells, as well as numbers of HIV-1 RNA copies per milliliter plasma were measured. RESULTS: Sequential analysis of VL, IC load on CD4(+) blood lymphocytes and CD4 counts showed that an increasing VL was not associated with CD4 depletion, when the proportion of IC-coated circulating CD4(+) blood lymphocytes remained stable. When, CD4 counts and IC load were analyzed during corresponding intervals of retroviral replication in the three patient groups, a higher VL was associated with lower CD4 counts only when the IC load (IgG or gp120/IgG) on CD4(+) lymphocytes was higher as well. CONCLUSION: These data suggest that HIV regulates and mediates CD4 depletion in part by the induction of autoreactive ICs against CD4(+) lymphocytes, especially complement-fixing autoreactive IgG and gp120/IgG complexes.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , HIV Infections/immunology , HIV-1/immunology , Hemophilia A/immunology , CD4 Antigens/immunology , CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/metabolism , Cell Count , Humans , Retrospective Studies , Viral Load
SELECTION OF CITATIONS
SEARCH DETAIL
...