Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
1.
Transpl Infect Dis ; 17(1): 25-32, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25572799

ABSTRACT

BACKGROUND: Natural immunity against cytomegalovirus (CMV) can control virus replication after solid organ transplantation; however, it is not known which components of the adaptive immune system mediate this protection. We investigated whether this protection requires human leukocyte antigen (HLA) matching between donor and recipient by exploiting the fact that, unlike transplantation of other solid organs, liver transplantation does not require HLA matching, but some donor and recipient pairs may nevertheless be matched by chance. METHODS: To further investigate this immune control, we determined whether chance HLA matching between donor (D) and recipient (R) in liver transplants affected a range of viral replication parameters. RESULTS: In total, 274 liver transplant recipients were stratified according to matches at the HLA A, HLA B, and HLA DR loci. The incidence of CMV viremia, kinetics of replication, and peak viral load were similar between the HLA matched and mismatched patients in the D+/R+ and D-/R+ transplant groups. D+/R- transplants with 1 or 2 mismatches at the HLA DR locus had a higher incidence of CMV viremia >3000 genomes/mL blood compared to patients matched at this locus (78% vs. 17%; P = 0.01). Evidence was seen that matching at the HLA A locus had a small effect on peak viral loads in D+/R- patients, with median peak loads of 3540 and 14,706 genomes/mL in the 0 and combined (1 and 2) mismatch groups, respectively (P = 0.03). CONCLUSION: Overall, our data indicate that, in the setting of liver transplantation, prevention of CMV infection and control of CMV replication by adaptive immunity is minimally influenced by HLA matching of the donor and recipient. Our data raise questions about immune control of CMV in the liver and also about the cells in which the virus is amplified to give rise to CMV viremia.


Subject(s)
Adaptive Immunity , Cytomegalovirus Infections/immunology , Cytomegalovirus/physiology , HLA Antigens/immunology , Liver Transplantation/adverse effects , Adult , Cytomegalovirus Infections/prevention & control , Female , Humans , Male , Middle Aged , Tissue Donors , Transplant Recipients , Virus Replication
2.
Am J Transplant ; 13 Suppl 3: 79-86; quiz 86, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23347216

ABSTRACT

Over the last few years there has been an impressive increase in the virological and immunological tools available to detect both human herpesvirus (HHV) and immune control of replication post-solid organ transplantation. This has allowed a greater appreciation of pathogenesis, studies to be designed to evaluate potential vaccines, new approaches adopted for antiviral deployment and the success of interventions to be judged. This chapter aims to summarize the state-of-the-art in vaccine development and look forward to the role that vaccines, immune monitoring, viral kinetics and new antiherpesvirus agents may play in the future management of HHV infections after transplantation.


Subject(s)
Cytomegalovirus Infections/complications , Herpesviridae Infections/complications , Herpesvirus Vaccines/therapeutic use , Immunotherapy, Adoptive/methods , Organ Transplantation/adverse effects , Animals , Antiviral Agents/therapeutic use , Cytomegalovirus Infections/diagnosis , Cytomegalovirus Infections/prevention & control , Herpesviridae Infections/diagnosis , Herpesviridae Infections/prevention & control , Humans , Immunotherapy/methods , Postoperative Complications
3.
Am J Transplant ; 12(9): 2457-64, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22594993

ABSTRACT

After allotransplantation, cytomegalovirus (CMV) may be transmitted from the donor organ, giving rise to primary infection in a CMV negative recipient or reinfection in one who is CMV positive. In addition, latent CMV may reactivate in a CMV positive recipient. In this study, serial blood samples from 689 kidney or liver transplant recipients were tested for CMV DNA by quantitative PCR. CMV was managed using preemptive antiviral therapy and no patient received antiviral prophylaxis. Dynamic and quantitative measures of viremia and treatment were assessed. Median peak viral load, duration of viremia and duration of treatment were highest during primary infection, followed by reinfection then reactivation. In patients who experienced a second episode of viremia, the viral replication rate was significantly slower than in the first episode. Our data provide a clear demonstration of the immune control of CMV in immunosuppressed patients and emphasize the effectiveness of the preemptive approach for prevention of CMV syndrome and end organ disease. Overall, our findings provide quantitative biomarkers which can be used in pharmacodynamic assessments of the ability of novel CMV vaccines or antiviral drugs to reduce or even interrupt such transmission.


Subject(s)
Cytomegalovirus/physiology , Organ Transplantation , Virus Replication/drug effects , Biomarkers , Humans , Immunosuppressive Agents/administration & dosage , Polymerase Chain Reaction , Viral Load
4.
J Exp Med ; 190(2): 177-82, 1999 Jul 19.
Article in English | MEDLINE | ID: mdl-10432281

ABSTRACT

Cytomegalovirus (CMV) is generally described as a slowly replicating virus. During studies of immunocompromised patients, we observed rapid changes in the quantity of CMV DNA present in serial blood samples by quantitative-competitive polymerase chain reaction commensurate with a doubling time of <2 d. To further investigate the dynamics of replication in vivo, patients in three distinct situations were studied in detail: (a) those receiving intravenous ganciclovir; (b) those in whom ganciclovir-resistant strains appeared during long-term therapy; and (c) those in whom ganciclovir-resistant strains disappeared with alternative drug therapy. In all cases, it was possible to provide accurate estimates of the doubling time of CMV and its half-life of disappearance after antiviral chemotherapy. The results from all three approaches demonstrated that the doubling time/half-life of CMV in blood is approximately 1 d when frequent samples are collected. These results show that CMV DNA replication in vivo is a highly dynamic process. We conclude that the reputation of CMV as a slowly replicating virus based on the time taken to produce cytopathic effects in vitro is unwarranted. These findings have implications for the potency, dose, and duration of antiviral chemotherapy needed for the effective treatment of this important human pathogen.


Subject(s)
Cytomegalovirus Infections/virology , Cytomegalovirus/physiology , Virus Replication/physiology , Antiviral Agents/therapeutic use , Base Sequence , Cytomegalovirus/drug effects , Cytomegalovirus Infections/drug therapy , DNA Primers/genetics , DNA Replication/drug effects , DNA Replication/physiology , DNA, Viral/blood , DNA, Viral/genetics , Drug Resistance, Microbial , Ganciclovir/therapeutic use , Genes, Viral , Humans , Kinetics , Mutation , Phosphotransferases (Alcohol Group Acceptor)/genetics , Virus Replication/drug effects
5.
J Exp Med ; 193(7): 847-54, 2001 Apr 02.
Article in English | MEDLINE | ID: mdl-11283157

ABSTRACT

Using patient data from a unique single source outbreak of hepatitis B virus (HBV) infection, we have characterized the kinetics of acute HBV infection by monitoring viral turnover in the serum during the late incubation and clinical phases of the disease in humans. HBV replicates rapidly with minimally estimated doubling times ranging between 2.2 and 5.8 d (mean 3.7 +/- 1.5 d). After a peak viral load in serum of nearly 10(10) HBV DNA copies/ml is attained, clearance of HBV DNA follows a two or three phase decay pattern with an initial rapid decline characterized by mean half-life (t(1/2)) of 3.7 +/- 1.2 d, similar to the t(1/2) observed in the noncytolytic clearance of covalently closed circular DNA for other hepadnaviruses. The final phase of virion clearance occurs at a variable rate (t(1/2) of 4.8 to 284 d) and may relate to the rate of loss of infected hepatocytes. Free virus has a mean t(1/2) of at most 1.2 +/- 0.6 d. We estimate a peak HBV production rate of at least 10(13) virions/day and a maximum production rate of an infected hepatocyte of 200-1,000 virions/day, on average. At this peak rate of virion production we estimate that every possible single and most double mutations would be created each day.


Subject(s)
DNA, Viral/blood , Hepatitis B virus/growth & development , Hepatitis B/blood , Acute Disease , DNA, Circular/metabolism , Disease Outbreaks , Half-Life , Hepatitis B/epidemiology , Humans , Kinetics , Liver/virology , Virion/growth & development
6.
Am J Transplant ; 8(12): 2590-9, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18853954

ABSTRACT

To determine whether polyfunctional CD4+ T-cell responses coupled with CD8+ T-cell responses against human cytomegalovirus (HCMV) are key to the control of HCMV replication we prospectively analyzed 29 liver transplant recipients for CD4+ T-cell responses against soluble HCMV antigen, pp65 and IE1 proteins, CD8+ T-cell responses against pp65 and IE1 proteins and a range of T helper (Th) 1 and Th2 cytokines. Eleven patients (38%) developed HCMV DNAemia at a median of 21 days post-liver transplantation (range 17-31 days). There was a significantly lower frequency and absolute number of total HCMV CD4+ T cells producing IFNgamma, IFNgamma+IL2 and IL2 and pp65-CD8+ T cells producing IFNgamma in patients with DNAemia. The quantities of Th1 and Th2 cytokines present during the first 20 days posttransplant were not predictive of DNAemia. Cut-off levels during the first 20 days posttransplant of 0.1% of lysate stimulated CD4+ T cells producing IL2, and pp65-CD8+ T cells producing IFNgamma above 0.4% had positive and negative predictive values for DNAemia of 54% and 100% and 50% and 92%, respectively. Measuring polyfunctional CD4+ T cells against HCMV early posttransplant may allow targeted intervention to minimize the occurrence and acute and long-term consequences of HCMV replication.


Subject(s)
CD4-Positive T-Lymphocytes/physiology , CD8-Positive T-Lymphocytes/physiology , Cytomegalovirus/physiology , Liver Transplantation/physiology , Phosphoproteins/physiology , Viral Matrix Proteins/physiology , Virus Replication/physiology , Adult , Aged , Antigens, Viral/metabolism , DNA, Viral/blood , Female , Humans , Immediate-Early Proteins/metabolism , Interferon-gamma/metabolism , Interleukin-2/metabolism , Linear Models , Male , Middle Aged , Prospective Studies , ROC Curve
7.
Am J Transplant ; 8(5): 990-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18325078

ABSTRACT

Human cytomegalovirus (HCMV) remains an important cause of morbidity after allotransplantation, causing a range of direct effects including hepatitis, pneumonitis, enteritis and retinitis. A dominant risk factor for HCMV disease is high level viral replication in blood but it remains unexplained why only a subset of patients develop such diseases. In this detailed study of 25 renal transplant recipients, we show that functional impairment of HCMV specific CD8 T cells in the production of interferon gamma was associated with a 14-fold increased risk of progression to high level replication. The CD8 T-cell impairment persisted during the period of high level replication and was more prominent in patients above 40 years of age (odds ratio = 1.37, p = 0.01) and was also evident in dialysis patients. Threshold levels of functional impairment were associated with an increased risk of future HCMV replication and there was a direct relationship between the functional capacity of HCMV ppUL83 CD8 T cells and HCMV load (R(2)= 0.83). These results help to explain why a subset of seropositive individuals develop HCMV replication and are at risk of end-organ disease and may facilitate the early identification of individuals who would benefit from targeted anti-HCMV therapy after renal transplantation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Kidney Transplantation/immunology , Antiviral Agents/therapeutic use , Cytomegalovirus/genetics , Cytomegalovirus/physiology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/epidemiology , DNA, Viral/genetics , DNA, Viral/isolation & purification , Female , Follow-Up Studies , Ganciclovir/therapeutic use , Humans , Interferon-gamma/blood , Kidney Transplantation/adverse effects , Male , Polymerase Chain Reaction , Postoperative Complications/epidemiology , Postoperative Complications/virology , Prospective Studies , Virus Replication
9.
J Clin Virol ; 82: 152-158, 2016 09.
Article in English | MEDLINE | ID: mdl-27500364

ABSTRACT

BACKGROUND: Cytomegalovirus (CMV) is the most prevalent congenital infection in developed countries. A significant number of infected infants develop long-term neurodevelopmental and hearing impairment irrespective of whether disease is detectable at birth. Studies of viral load and replication dynamics have informed the treatment of CMV in adult populations but no similar data exist in neonates. OBJECTIVES: To study CMV virus kinetics in different body fluids of babies treated for congenital infection. STUDY DESIGN: CMV virus load was sequentially analyzed in blood, urine and saliva in 17 babies treated for symptomatic congenital CMV infection. RESULTS: Virus was detectable in the urine and saliva of all babies at baseline but in only 15/17 in blood. At the end of 6 weeks of antiviral treatment CMV remained detectable in 9/14 blood samples, 9/12 urine samples and 4/7 salivary swabs. Median half-life (T1/2) of virus decline in blood was 2.4 days (IQR 1.9-3.3) and basic reproductive number (Ro) was 2.3. Although T1/2 values were similar in urine and saliva to those observed in blood, virus dynamics differed both during and after treatment. CONCLUSIONS: T1/2 and Ro in blood in this group of neonates were similar to values derived from studies of immunocompromised adults. The persistent viremia observed in treated neonates cannot therefore be adequately explained by the virus dynamics early in treatment. The different dynamics exhibited in blood and urine suggests that studying changes in distinct body compartments may assist in further understanding long-term manifestations of disease.


Subject(s)
Cytomegalovirus Infections/congenital , Cytomegalovirus Infections/virology , Cytomegalovirus/isolation & purification , Viral Load , Blood/virology , Cytomegalovirus/physiology , Humans , Infant , Infant, Newborn , Saliva/virology , Time Factors , Urine/virology , Virus Replication
10.
Mol Immunol ; 35(9): 503-12, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9809578

ABSTRACT

Factor I is a five-domain plasma serine protease which is essential for the regulation of the complement system. In order to express this, the factor I coding sequence was cloned into a recombinant baculovirus system, which was used to infect Trichoplusia ni cells. Using the native factor I leader sequence, recombinant factor I (rFI) was secreted into the culture medium. Purified rFI was recognised by polyclonal antisera and by the factor I-specific monoclonal antibody MRC-OX21. SDS PAGE showed that rFI was processed into two chains with molecular weights of 48,000 and 36,000. Amino acid sequence analysis showed that the N-terminal sequences of the rFI chains were the same as those of serum-derived factor I (sFI), confirming that processing was correct. Since both molecular weights were less than those observed for sFI, this is attributed to the replacement of complex-type oligosaccharides by high mannose ones in rFI. C3(NH,) cleavage assays showed that rFI had 55% the activity of sFI. Circular dichroism and Fourier transform infrared spectroscopy showed that the protein folding of rFI and sFI were very similar. Both had a secondary structure low in alpha-helix and high in beta-sheet, as expected from crystal structure and multiple sequence alignment analyses. It is inferred that the reduced activity of rFI is attributable to its changed glycosylation. The availability of rFI and structures for the domains in factor I makes possible new approaches to determine the molecular basis of its interactions with factor H and C3b.


Subject(s)
Complement Factor I/biosynthesis , Animals , Baculoviridae/genetics , Circular Dichroism , Cloning, Molecular , Complement Factor I/chemistry , Complement Factor I/genetics , Glycosylation , Humans , Moths/cytology , Protein Processing, Post-Translational , Protein Structure, Secondary , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Spectroscopy, Fourier Transform Infrared
11.
AIDS ; 5(5): 485-9, 1991 May.
Article in English | MEDLINE | ID: mdl-1677807

ABSTRACT

A new mechanism is proposed for the apparent breakthrough of HIV that occurs approximately 6 months after the commencement of therapy with zidovudine (AZT). Using a simple mathematical model of the interacting population dynamics of HIV and its major host cell in the circulation (the CD4+ lymphocyte), predicted patterns of HIV plasma viraemia in the weeks following treatment with zidovudine are generated. These are in close agreement with observed patterns despite the fact that the model contains no mechanisms for the development of drug-resistant strains of virus. It is suggested that the patterns of viral abundance observed during the first 6 months after treatment may be the result of non-linearities in the interactions between HIV and CD4+ cells, and that it is only after the first post-treatment burst of viral production that drug resistance plays an important role.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , CD4-Positive T-Lymphocytes/microbiology , HIV Infections/drug therapy , HIV-1/drug effects , Zidovudine/therapeutic use , AIDS-Related Complex/drug therapy , AIDS-Related Complex/microbiology , Acquired Immunodeficiency Syndrome/microbiology , CD4-Positive T-Lymphocytes/drug effects , Drug Resistance, Microbial , HIV Infections/microbiology , HIV-1/growth & development , Humans , Leukocyte Count , Models, Theoretical , Time Factors , Viremia
12.
AIDS ; 13(10): 1203-6, 1999 Jul 09.
Article in English | MEDLINE | ID: mdl-10416523

ABSTRACT

OBJECTIVE: To determine the effect of highly active antiretroviral therapy (HAART) on cytomegalovirus (CMV) viraemia and retinitis in patients at high risk of disease. DESIGN: Sixteen patients with CMV viraemia, but no evidence of end organ disease at the time of first receipt of HAART including a protease inhibitor, were studied. No patient had ever received specific anti-CMV therapy. METHODS: CMV load in blood was measured using quantitative competitive PCR at baseline and for a median follow-up of 21 months. Regular ophthalmological screening for retinitis was conducted throughout the study period. RESULTS: All 16 patients became CMV negative by PCR following the commencement of HAART. CMV loads prior to treatment ranged from 2.0 x 10(3) to 4.1 x 10(6) copies/ml (median, 7.6 x 10(4) copies/ml). The median time to becoming PCR negative was 13.5 weeks (range, 5-40 weeks). Fourteen patients remained CMV negative throughout follow-up. CMV viraemia recurred in two patients; these individuals were indistinguishable with respect to either baseline parameters or response to antiretroviral therapy. None of the 16 patients developed CMV retinitis. CONCLUSIONS: HAART including a protease inhibitor can result in the complete suppression of CMV viraemia, an effect not previously observed in HIV-infected patients in the absence of specific anti-CMV therapy. This response correlated with protection against CMV retinitis in a group of patients at high risk of development of disease. These results help to explain why the natural history of CMV disease has altered since the introduction of such therapeutic regimens.


Subject(s)
AIDS-Related Opportunistic Infections/drug therapy , Anti-HIV Agents/therapeutic use , Cytomegalovirus Infections/drug therapy , Cytomegalovirus/drug effects , Viremia/drug therapy , AIDS-Related Opportunistic Infections/virology , Adult , CD4 Lymphocyte Count , Cytomegalovirus/physiology , Cytomegalovirus Infections/virology , Cytomegalovirus Retinitis/prevention & control , Drug Therapy, Combination , Female , HIV Protease Inhibitors/therapeutic use , HIV-1/physiology , Humans , Male , Middle Aged , Polymerase Chain Reaction/methods , RNA, Viral/blood , Reverse Transcriptase Inhibitors/therapeutic use , Viremia/virology
13.
AIDS ; 9(7): 675-83, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7546410

ABSTRACT

OBJECTIVE: To investigate the phylogenetic relationship of HIV-1 proviral long terminal repeat (LTR) variants present in postmortem samples of lymph node, spleen, lung, dorsal root ganglion and spinal cord as well as in the peripheral blood of an HIV-1-infected patient dying with AIDS. DESIGN AND METHODS: Postmortem tissues were studied by a combination of histology, cell culture and molecular analyses. The patient had a stable CD4 count of 10 x 10(6)/I during the 12 months preceding death. A 540 base-pair fragment of the LTR including U3/R/U5 was amplified using polymerase chain reaction on proviral DNA from the five postmortem tissues and peripheral blood mononuclear cells obtained 2 months prior to death. The population of viral variants was determined by sequencing at least five plasmid clones of the amplicons. The relationship between the variants present in different body sites was investigated using molecular phylogeny methods. RESULTS: HIV-1 was present in all organs analysed and correlated with the presence of abnormal histology. Genetic variation leading to divergence from the consensus sequence was more frequently present in characterized transcription factor binding sites within the LTR (P < 0.0001) although the HIV-1 LTR quasispecies in the different body sites showed similar, relatively low levels of divergence (intra-organ median heterogeneity ranging from 0.0094 to 0.017). Phylogenetic analysis showed that the spinal cord and dorsal root ganglion harboured an LTR population genetically distinct from that present in other organs and more closely related to a previously characterized neurotropic strain of HIV (strain JRcsf). CONCLUSION: The independent clustering of HIV-1 LTR variants present in spinal cord and dorsal root ganglion shows that HIV-1 LTR evolution can occur in a compartmentalized fashion. The data show that the LTR is an important region to analyse in sequence variation studies of HIV since it may play a role in nervous tissue adaptation of HIV-1 and neuropathogenicity. Outgrowth of HIV-1 LTR variants that are most fit for the utilization of tissue-specific transcription factors can occur in the nervous tissue.


Subject(s)
Acquired Immunodeficiency Syndrome/genetics , HIV Long Terminal Repeat , HIV-1/genetics , Acquired Immunodeficiency Syndrome/blood , Base Sequence , Ganglia, Spinal/virology , Genetic Heterogeneity , HIV-1/classification , Humans , Lung/virology , Lymph Nodes/virology , Male , Molecular Sequence Data , Phylogeny , Polymerase Chain Reaction , Sequence Alignment , Spinal Cord/virology
14.
AIDS ; 10 Suppl 1: S37-41, 1996 Nov.
Article in English | MEDLINE | ID: mdl-8970675

ABSTRACT

BACKGROUND: More than 90% of patients with HIV have been infected at some time with cytomegalovirus (CMV) and up to 40% of those with advanced HIV will develop CMV disease. The incidence of CMV disease is increasing but the prognosis for the patient remains poor. MONITORING FOR CMV: It is therefore important to monitor patients with low CD4+ counts in order to identify those most at risk of developing CMV disease and to treat them before the disease becomes established. Polymerase chain reaction (PCR) is probably the most effective and sensitive method of detecting CMV and a positive result is predictive for development of CMV disease; more than 80% of patients with CMV retinitis are CMV PCR-positive at the time of diagnosis. PCR can also detect the presence of CMV up to 14 months before the development of retinitis. TREATMENT OF CMV RETINITIS: In patients with detectable CMV, but no evidence of active infection, pre-emptive treatment with ganciclovir or valaciclovir has been shown to reduce the risk of developing retinitis in these high-risk patients. Such oral therapy, which is generally better tolerated than intravenous therapy and results in a better quality of life for the patient, is likely to be more effective at this stage whilst viral loads are low. CONCLUSIONS: CMV PCR can be used to prospectively monitor patients in order to identify those most at risk of developing CMV retinitis. If CMV infection is diagnosed early, while viral loads are still low, pre-emptive oral therapy can be instituted which will reduce the chances of developing retinitis in those patients most at risk.


Subject(s)
AIDS-Related Opportunistic Infections/diagnosis , Cytomegalovirus Infections/complications , AIDS-Related Opportunistic Infections/drug therapy , AIDS-Related Opportunistic Infections/virology , Acyclovir/administration & dosage , Acyclovir/analogs & derivatives , Administration, Oral , Antiviral Agents/administration & dosage , Cytomegalovirus/genetics , Cytomegalovirus/isolation & purification , Cytomegalovirus Infections/diagnosis , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Retinitis/complications , Cytomegalovirus Retinitis/drug therapy , Cytomegalovirus Retinitis/prevention & control , Ganciclovir/administration & dosage , Humans , Polymerase Chain Reaction , Risk Factors , Valacyclovir , Valine/administration & dosage , Valine/analogs & derivatives
15.
AIDS ; 11(7): 889-93, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9189214

ABSTRACT

BACKGROUND: Cytomegalovirus (CMV) disease is a major cause of morbidity in patients with HIV infection. Despite treatment, CMV retinitis causes substantial visual loss, especially in patients with CD4 cell counts below 50 x 10(6)/l. Although routine ophthalmological screening of these patients has been recommended, no controlled trials have evaluated how frequently it should be performed. The aim of this study was to assess whether CMV polymerase chain reaction (PCR) results could direct ophthalmological screening to patients at high risk of CMV retinitis. METHODS: In a prospective study of HIV-positive patients with CD4 cell counts below 50 x 10(6)/l, CMV viraemia was detected by qualitative PCR of whole blood. Patients who were CMV PCR-viraemic were allocated to monthly virological and ophthalmological follow-up; patients who were PCR-negative received 3-monthly virological and ophthalmological follow-up. CMV viral load was determined in all CMV-positive samples using a quantitative competitive PCR. RESULTS: Nineteen out of 97 patients developed CMV disease over the first 12 months of the study. Sixteen (59%) out of 27 patients who were CMV-positive developed disease compared with three (4%) out of 70 of patients who were PCR-negative (P = 0.0001). A positive CMV PCR result was significantly associated with the development of disease (P = 0.0001), with a relative hazard of 20.15 [95% confidence interval (CI), 5.80-69.98]. Median CMV viral load was significantly higher in those individuals who went on to develop CMV disease (P = 0.02). In PCR-positive patients, each 0.25 log10 increase in viral load increased the risk of disease (relative hazard, 1.37; 95% CI, 1.15-1.63; P = 0.0004). CONCLUSIONS: CMV PCR predicts the development of CMV disease and can be used to target ophthalmological resources to those patients at highest risk of retinitis. Asymptomatic patients who are PCR-positive represent a high-risk group in whom controlled trials of pre-emptive therapy could be conducted.


Subject(s)
AIDS-Related Opportunistic Infections/diagnosis , Cytomegalovirus Retinitis/diagnosis , Polymerase Chain Reaction , Viremia/diagnosis , AIDS-Related Opportunistic Infections/virology , Adult , Cytomegalovirus Retinitis/complications , Follow-Up Studies , Humans , Middle Aged , Predictive Value of Tests , Prospective Studies , Risk Factors , Viral Load , Viremia/complications , Viremia/virology
16.
AIDS ; 12(6): 605-11, 1998 Apr 16.
Article in English | MEDLINE | ID: mdl-9583600

ABSTRACT

OBJECTIVES: To determine whether recurrence of polymerase chain reaction (PCR) viraemia during maintenance ganciclovir for cytomegalovirus (CMV) retinitis correlates with (i) CMV disease at a new anatomical site, (ii) progression of the presenting retinitis, or (iii) acquisition of genetic changes in gene UL97 associated with resistance to ganciclovir. DESIGN: A previously described cohort of 45 patients presenting with first episode retinitis was followed clinically using ophthalmoscopy and serial tests for PCR viraemia for a median of 7 months. CMV viral load and genetic markers of ganciclovir resistance were measured in PCR-positive samples. METHODS: PCR amplification of the glycoprotein B region of CMV and quantitative competitive PCR assays were employed. Genetic changes in UL97 were identified by sequencing/point mutation assay. RESULTS: PCR viraemia correlated significantly with new episodes of CMV disease (P=0.011) and a trend was seen for the association with progression of retinitis (P=0.07). Amongst the 14 patients PCR-positive during maintenance ganciclovir, 10 (71%) had genetic markers of resistance. None of these patients became PCR-negative in blood after reinduction ganciclovir therapy compared with three out of four without markers of resistance (P=0.022). CONCLUSIONS: CMV PCR viraemia correlated strongly with the development of new episodes of CMV disease. Most patients with progression of retinitis remained PCR-negative in blood, consistent with therapeutic failure due to poor intraocular penetration of ganciclovir. However, the minority who were PCR-positive in blood may have reinfected their eye, and frequently had markers of ganciclovir resistance. The implications of these findings for the management of patients with CMV disease are discussed.


Subject(s)
Antiviral Agents/therapeutic use , Cytomegalovirus Retinitis/drug therapy , Cytomegalovirus Retinitis/virology , Cytomegalovirus/genetics , Ganciclovir/therapeutic use , Viremia/virology , Cohort Studies , Cytomegalovirus/isolation & purification , Cytomegalovirus Infections/virology , DNA Mutational Analysis , DNA, Viral/blood , Disease Progression , Drug Resistance, Microbial/genetics , Genotype , Humans , Male , Phosphotransferases (Alcohol Group Acceptor)/genetics , Point Mutation/genetics , Polymerase Chain Reaction/methods , Prospective Studies , Recurrence , Viral Envelope Proteins/genetics
17.
AIDS ; 14(9): 1163-70, 2000 Jun 16.
Article in English | MEDLINE | ID: mdl-10894280

ABSTRACT

OBJECTIVE: To determine the effect of highly active antiretroviral therapy (HAART) on the natural history of cytomegalovirus (CMV) retinitis. DESIGN AND PARTICIPANTS: Retrospective analysis of 103 consecutive patients diagnosed with CMV retinitis between 1990 and 1998. SETTING: Specialist HIV medicine department of a London hospital. MAIN OUTCOME MEASURES: Incidence of CMV retinitis, time to death following diagnosis, episodes of progression, incidence of inflammatory complications. The date of first use of HAART was January 1995. Data were censored on 30 June 1998. RESULTS: The incidence of CMV retinitis has declined dramatically following the introduction of HAART. Survival following CMV retinitis increased from a median of 0.65 years prior to 1995 to a median of 1.07 years after this date (P = 0.004). In multivariate analyses HAART was independently associated with improved survival (P = 0.02) and the association with year of diagnosis was no longer significant, suggesting that this effect is predominantly due to HAART. None of the patients receiving HAART experienced progression after 6 months of treatment. Complications of retinitis such as retinal detachment, uveitis and optic atrophy occurred in 39% of patients. The rare inflammatory complications of vitritis and cystoid macular oedema occurred only in recipients of HAART. CONCLUSIONS: The introduction of HAART has had a major impact on the natural history of CMV retinitis with improved survival time and decreased risk of progression following diagnosis. However, immune reconstitution may be associated with inflammatory complications which can result in significant visual loss in the absence of active CMV disease.


Subject(s)
AIDS-Related Opportunistic Infections/physiopathology , Anti-HIV Agents/therapeutic use , Antiretroviral Therapy, Highly Active , Cytomegalovirus Retinitis/physiopathology , HIV Infections/drug therapy , AIDS-Related Opportunistic Infections/epidemiology , AIDS-Related Opportunistic Infections/prevention & control , Adult , Cohort Studies , Cytomegalovirus Retinitis/epidemiology , Cytomegalovirus Retinitis/prevention & control , Disease Progression , Female , HIV Infections/mortality , Humans , Incidence , Inflammation , London/epidemiology , Male , Middle Aged , Retrospective Studies , Survival Rate , Time Factors
18.
FEBS Lett ; 371(2): 199-203, 1995 Sep 04.
Article in English | MEDLINE | ID: mdl-7672128

ABSTRACT

Low density lipoprotein receptor domains (LDLrs) represent a large cell surface receptor superfamily of consensus length 39 residues. Alignment of 194 sequences indicated highly conserved Cys and Asp/Glu residues, and a consensus secondary structure with three beta-strands was predicted. Sequence threading against known protein folds indicated consistency with small beta-sheet proteins. Complement factor I contains two LDLrs, and the second of these was successfully expressed using a bacterial pGEX system. FT-IR spectroscopy on this indicated a small amount of beta-sheet together with turns and loops. LDLr is proposed to have a beta-sheet structure in which the five biologically important Asp/Glu residues are located on an exposed loop.


Subject(s)
Complement Factor I/chemistry , Protein Structure, Secondary , Receptors, LDL/chemistry , Amino Acid Sequence , Circular Dichroism , Consensus Sequence , Humans , Molecular Sequence Data , Protein Folding , Sequence Alignment , Spectrometry, Fluorescence , Spectroscopy, Fourier Transform Infrared
19.
Antivir Ther ; 3(4): 239-42, 1998.
Article in English | MEDLINE | ID: mdl-10682146

ABSTRACT

Clinical resistance of cytomegalovirus (CMV) against the currently licensed antiviral drugs is becoming an increasingly recognized problem. This review focuses on the molecular basis of resistance and describes mutations in the UL54 DNA polymerase leading to resistance against cidofovir, foscarnet and ganciclovir. The review highlights two important developments in our appreciation of resistance. Firstly, the use of more rapid molecular based assays to detect genotypic resistance and secondly, the relationship between resistance profiles in multiple organ systems of the same host. Finally, the changing face of CMV disease in the era of highly active antiviral chemotherapy is considered with respect to its impact on the frequency of CMV resistance in the clinic.


Subject(s)
Antiviral Agents/therapeutic use , Cytomegalovirus Infections/drug therapy , Organophosphonates , Cidofovir , Cytosine/analogs & derivatives , Cytosine/therapeutic use , Drug Resistance , Foscarnet/therapeutic use , Ganciclovir/therapeutic use , Humans , Mutation , Organophosphorus Compounds/therapeutic use
20.
Transplantation ; 71(11 Suppl): SS24-30, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11583485

ABSTRACT

Viral infections, particularly those involving HCMV, are an important complication of renal transplantation. Transplantation protocols and treatment regimens that increase HCMV infection and disease may promote the development of CRAD and impair long-term renal allograft survival. Investigators are beginning to illuminate the mechanisms by which HCMV infection may cause chronic rejection in general and transplant vascular sclerosis in particular. Migration and proliferation of SMCs within the intimal layer of blood vessels is an important component of transplant vascular sclerosis, and HCMV appears to facilitate both of these processes. Current management strategies for HCMV focus on prevention, either using a focal preemptive therapeutic approach or by administering antiviral therapies to all or at-risk patients.


Subject(s)
Cytomegalovirus Infections/complications , Graft Rejection/virology , Kidney Transplantation , Virus Diseases/complications , Antiviral Agents/therapeutic use , Chronic Disease , Cytomegalovirus Infections/prevention & control , Graft Rejection/pathology , Humans , Muscle, Smooth, Vascular/pathology
SELECTION OF CITATIONS
SEARCH DETAIL