1
|
Tsai YC, Tsai TF. Oral disease-modifying antirheumatic drugs and immunosuppressants with antiviral potential, including SARS-CoV-2 infection: a review. Ther Adv Musculoskelet Dis 2020; 12:1759720X20947296. [PMID: 32952617 PMCID: PMC7476354 DOI: 10.1177/1759720x20947296] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/02/2020] [Indexed: 12/23/2022] Open
Abstract
There have been several episodes of viral infection evolving into epidemics in recent decades, and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the latest example. Its high infectivity and moderate mortality have resulted in an urgent need to find an effective treatment modality. Although the category of immunosuppressive drugs usually poses a risk of infection due to interference of the immune system, some of them have been found to exert antiviral properties and are already used in daily practice. Recently, hydroxychloroquine and baricitinib have been proposed as potential drugs for SARS-CoV-2. In fact, there are other immunosuppressants known with antiviral activities, including cyclosporine A, hydroxyurea, minocycline, mycophenolic acid, mycophenolate mofetil, leflunomide, tofacitinib, and thalidomide. The inherent antiviral activity could be a treatment choice for patients with coexisting rheumatological disorders and infections. Clinical evidence, their possible mode of actions and spectrum of antiviral activities are included in this review article. LAY SUMMARY Immunosuppressants often raise the concern of infection risks, especially for patients with underlying immune disorders. However, some disease-modifying antirheumatic drugs (DMARDs) with inherent antiviral activity would be a reasonable choice in the situation of concomitant viral infections and flare up of autoimmune diseases. This review covers DMARDs of treatment potential for SARS-CoV-2 in part I, and antiviral mechanisms plus trial evidence for viruses other than SARS-CoV-2 in part II.
Collapse
Affiliation(s)
- Y. C. Tsai
- Department of Dermatology, Far Eastern Memorial Hospital, New Taipei city, Taiwan
| | - T. F. Tsai
- Department of Dermatology, National Taiwan University Hospital and National Taiwan University College of Medicine, No. 7, Zhongshan S. Rd, Zhongzheng District, Taipei City 100, Taiwan
| |
Collapse
|
2
|
Lee ES, Heller MM, Kamangar F, Park K, Liao W, Koo J. Hydroxyurea for the Treatment of Psoriasis with an Emphasis on HIV-Infected Psoriasis Patients: A Review. ACTA ACUST UNITED AC 2018. [DOI: 10.1177/247553031117a00302] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Hydroxyurea is a drug that has been long forgotten for the treatment of psoriasis. In addition to its anti-psoriatic effects, it has also been shown to have antiviral effects. This dual effect makes it a drug that dermatologists may want to consider when treating psoriasis in HIV-infected patients. Currently, no studies are available that discuss the safety and efficacy of hydroxyurea in the treatment of psoriasis in this immunocompromised group; however, numerous reports discuss the safety and efficacy of hydroxyurea in psoriasis and HIV separately. This review suggests that hydroxyurea is generally safe and effective. The main risk involves the hematologic adverse events (anemia, leukopenia, thrombocytopenia, and macrocytosis), which appear to be dose dependent. Because of the common hematologic adverse events, hydroxyurea may be considered a viable therapeutic option for patients with generalized psoriasis that is inadequately responsive to other safer options, whether or not the patient is HIV positive.
Collapse
Affiliation(s)
- Eric S. Lee
- Department of Dermatology, Psoriasis and Skin Treatment Center, University of California, San Francisco, California
| | - Misha M. Heller
- Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Faranak Kamangar
- University of California, Davis School of Medicine, Sacramento, California
| | - Kelly Park
- Department of Dermatology, Psoriasis and Skin Treatment Center, University of California, San Francisco, California
| | - Wilson Liao
- Department of Dermatology, Psoriasis and Skin Treatment Center, University of California, San Francisco, California
| | - John Koo
- Department of Dermatology, Psoriasis and Skin Treatment Center, University of California, San Francisco, California
| |
Collapse
|
3
|
Fonteh P, Meyer D. In vitro reactivation of latent HIV-1 by cytostatic bis(thiosemicarbazonate) gold(III) complexes. BMC Infect Dis 2014; 14:680. [PMID: 25495419 PMCID: PMC4265357 DOI: 10.1186/s12879-014-0680-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 12/03/2014] [Indexed: 11/18/2022] Open
Abstract
Background A number of cytostatic agents have been investigated for the ability to reactivate latent viral reservoirs, which is a major prerequisite for the eradication of HIV-1 infection. Two cytostatic bis(thiosemicarbazonate) gold(III) complexes (designated 1 and 2) were tested for this potential in the U1 latency model of HIV-1 infection. Methods Cell viability in the presence or absence of 1 and 2 was determined using a tetrazolium dye and evidence of reactivation was assessed by p24 antigen capture following exposure to a latency stimulant, phorbol myristate acetate (PMA) and or test compounds. The latency reactivation mechanism was explored by determining the effect of the complexes on protein kinase C (PKC), histone deacetylases (HDAC) and proinflammatory cytokine production. Results The CC50 of the complexes in U1 cells were 0.53 ± 0.12 μM for 1 and 1.0 ± 0.4 μM for 2. In the absence of PMA and at non toxic concentrations of 0.2 and 0.5 μM, 1 and 2 significantly (p ≤ 0.02) reactivated virus in U1 cells by 2.7 and 2.3 fold respectively. In comparison, a 2.6 fold increase (p = 0.03) in viral reactivation was observed for hydroxyurea (HU), which was used as a cytostatic and latent HIV reactivation control. Viral reactivation was absent for the complexes during co-stimulation with PMA indicating the lack of an additive effect between the chemicals as well as an absence of inhibition of PMA induced HIV reactivation but for HU inhibition of the stimulant’s activity was observed (p = 0.01). Complex 1 and 2 activated PKC activity by up to 32% (p < 0.05) but no significant inhibition of HDAC was observed. Increases in TNF-α levels suggested that the reactivation of virus by the complexes may have been due to contributions from the latter and the activation of PKC. Conclusion The ethyl group structural difference between 1 and 2 seems to influence bioactivity with lower active concentrations of 1, suggesting that further structural modifications should improve specificity. The cytostatic effect of 1 and 2 and now HIV reactivation from a U1 latency model is consistent with that of the cytostatic agent, HU. These findings suggest that the complexes have a potential dual (cytostatic and reactivation) role in viral “activation/elimination”. Electronic supplementary material The online version of this article (doi:10.1186/s12879-014-0680-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Debra Meyer
- Department of Biochemistry, Faculty of Natural and Agricultural Sciences, University of Pretoria, Hatfield Campus, Pretoria 0002, South Africa.
| |
Collapse
|
4
|
Beach LB, Rawson JM, Kim B, Patterson SE, Mansky LM. Novel inhibitors of human immunodeficiency virus type 2 infectivity. J Gen Virol 2014; 95:2778-2783. [PMID: 25103850 DOI: 10.1099/vir.0.069864-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Human immunodeficiency virus type 2 (HIV-2) infects about two million people worldwide. HIV-2 has fewer treatment options than HIV-1, yet may evolve drug resistance more quickly. We have analysed several novel drugs for anti-HIV-2 activity. It was observed that 5-azacytidine, clofarabine, gemcitabine and resveratrol have potent anti-HIV-2 activity. The EC50 values for 5-azacytidine, clofarabine and resveratrol were found to be significantly lower with HIV-2 than with HIV-1. A time-of-addition assay was used to analyse the ability of these drugs to interfere with HIV-2 replication. Reverse transcription was the likely target for antiretroviral activity. Taken together, several novel drugs have been discovered to have activity against HIV-2. Based upon their known activities, these drugs may elicit enhanced HIV-2 mutagenesis and therefore be useful for inducing HIV-2 lethal mutagenesis. In addition, the data are consistent with HIV-2 reverse transcriptase being more sensitive than HIV-1 reverse transcriptase to dNTP pool alterations.
Collapse
Affiliation(s)
- Lauren B Beach
- Molecular, Cellular, Developmental Biology & Genetics Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA.,Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Jonathan M Rawson
- Molecular, Cellular, Developmental Biology & Genetics Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA.,Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Baek Kim
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Steven E Patterson
- Center for Drug Design, University of Minnesota, Minneapolis, MN 55455, USA.,Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Louis M Mansky
- Center for Drug Design, University of Minnesota, Minneapolis, MN 55455, USA.,Molecular, Cellular, Developmental Biology & Genetics Graduate Program, University of Minnesota, Minneapolis, MN 55455, USA.,Institute for Molecular Virology, University of Minnesota, Minneapolis, MN 55455, USA.,Department of Diagnostic and Biological Sciences, School of Dentistry, University of Minnesota, Minneapolis, MN 55455, USA.,Department of Microbiology, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
5
|
Bonnac LF, Mansky LM, Patterson SE. Structure–Activity Relationships and Design of Viral Mutagens and Application to Lethal Mutagenesis. J Med Chem 2013; 56:9403-14. [DOI: 10.1021/jm400653j] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Laurent F. Bonnac
- Center for Drug Design, Academic
Health Center, University of Minnesota,
Minneapolis, Minnesota 55455, United States
| | - Louis M. Mansky
- Institute for Molecular Virology,
Academic Health Center, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Steven E. Patterson
- Center for Drug Design, Academic
Health Center, University of Minnesota,
Minneapolis, Minnesota 55455, United States
| |
Collapse
|
6
|
Abstract
PURPOSE OF REVIEW Interest in finding a potential 'cure' for HIV has taken on greater interest and urgency since the report of an individual who underwent allogeneic stem cell transplant from a CCR5 delta 32 homozygote donor after high-dose chemotherapy for acute myeloid leukemia. The potential role of cancer chemotherapy and other cancer-directed treatment approaches is discussed in the context of their potential role in helping to eliminate HIV from the infected host. RECENT FINDINGS Cancer chemotherapy and other cancer-targeted agents have been used successfully in treating a variety of malignancies in both HIV-infected and HIV-uninfected individuals. Lessons learned from these strategies may be of importance in helping to define more effective ways of controlling and eliminating HIV as well. Application of these anticancer strategies to patients with HIV are beginning to be explored and may help determine their potential usefulness in this disease as well. SUMMARY Although cytotoxic chemotherapy is a crude and not particularly effective way of removing HIV latently infected cells and tissue reservoirs, several new approaches to targeting and controlling cancer proliferation may be of value in HIV cure research and may one day help to end this disease.
Collapse
Affiliation(s)
- Ronald Mitsuyasu
- David Geffen School of Medicine at UCLA, UCLA Center for Clinical AIDS Research and Education (CARE Center), University of California, Los Angeles, CA 90035, USA.
| |
Collapse
|
7
|
Lori F, De Forni D, Katabira E, Baev D, Maserati R, Calarota SA, Cahn P, Testori M, Rakhmanova A, Stevens MR. VS411 reduced immune activation and HIV-1 RNA levels in 28 days: randomized proof-of-concept study for antiviral-hyperactivation limiting therapeutics. PLoS One 2012; 7:e47485. [PMID: 23094055 PMCID: PMC3477169 DOI: 10.1371/journal.pone.0047485] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 09/11/2012] [Indexed: 01/09/2023] Open
Abstract
Background A new class of antiretrovirals, AntiViral-HyperActivation Limiting Therapeutics (AV-HALTs), has been proposed as a disease-modifying therapy to both reduce Human Immunodeficiency Virus Type 1 (HIV-1) RNA levels and the excessive immune activation now recognized as the major driver of not only the continual loss of CD4+ T cells and progression to Acquired Immunodeficiency Syndrome (AIDS), but also of the emergence of both AIDS-defining and non-AIDS events that negatively impact upon morbidity and mortality despite successful (ie, fully suppressive) therapy. VS411, the first-in-class AV-HALT, combined low-dose, slow-release didanosine with low-dose hydroxycarbamide to accomplish both objectives with a favorable toxicity profile during short-term administration. Five dose combinations were administered as VS411 to test the AV-HALT Proof-of-Concept in HIV-1-infected subjects. Methods Multinational, double-blind, 28-day Phase 2a dose-ranging Proof-of-Concept study of antiviral activity, immunological parameters, safety, and genotypic resistance in 58 evaluable antiretroviral-naïve HIV-1-infected adults. Randomization and allocation to study arms were carried out by a central computer system. Results were analyzed by ANOVA, Kruskal-Wallis, ANCOVA, and two-tailed paired t tests. Results VS411 was well-tolerated, produced significant reductions of HIV-1 RNA levels, increased CD4+ T cell counts, and led to significant, rapid, unprecedented reductions of immune activation markers after 28 days despite incomplete viral suppression and without inhibiting HIV-1-specific immune responses. The didanosine 200 mg/HC 900 mg once-daily formulation demonstrated the greatest antiviral efficacy (HIV-1 RNA: −1.47 log10 copies/mL; CD4+ T cell count: +135 cells/mm3) and fewest adverse events. Conclusions VS411 successfully established the Proof-of-Concept that AV-HALTs can combine antiviral efficacy with rapid, potentially beneficial reductions in the excessive immune system activation associated with HIV-1 disease. Rapid reductions in markers of immune system hyperactivation and cellular proliferation were obtained despite the fact that VS411 did not attain maximal suppression of HIV RNA, suggesting this effect was due to the HALT component. Trial Registration ITEudraCT 2007-002460-98
Collapse
|
8
|
Treating HIV/AIDS by reducing immune system activation: the paradox of immune deficiency and immune hyperactivation. Curr Opin HIV AIDS 2012; 3:99-103. [PMID: 19372949 DOI: 10.1097/coh.0b013e3282f525cf] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
PURPOSE OF REVIEW To collect published evidence in support of a novel immune therapeutic approach to reduce the excess of immune activation that ultimately turns into immune deficiency in HIV/AIDS. RECENT FINDINGS A large body of evidence has been collected in support of the pathogenetic interpretation that prolonged immune overactivation induced by HIV during the course of chronic infection exhausts the immune system and leads to AIDS. Some groups are exploring the possibility of therapeutic interventions to limit the immune system overload. Cytostatic drugs appear promising candidates to achieve the goal, as they restrain cell proliferation without blocking it. At the same time, they do not affect cellular response to antigenic stimulation, and at appropriate dosages are not immune suppressive. SUMMARY Presently available antiretrovirals only partially reduce immune system activation during chronic HIV infection. Further clinical research is warranted to test cytostatic drugs to avert immune overactivation and prevent progression to AIDS.
Collapse
|
9
|
Fonteh PN, Keter FK, Meyer D. New bis(thiosemicarbazonate) gold(III) complexes inhibit HIV replication at cytostatic concentrations: potential for incorporation into virostatic cocktails. J Inorg Biochem 2011; 105:1173-80. [PMID: 21708102 DOI: 10.1016/j.jinorgbio.2011.05.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2011] [Revised: 05/13/2011] [Accepted: 05/18/2011] [Indexed: 10/18/2022]
Abstract
Four bis(thiosemicarbazonate)gold(III) complexes (1-4) with a general formula [Au(L)]Cl {L=L1, glyoxal-bis(N(4)-methylthiosemicarbazone); L2, glyoxal-bis(N(4)-ethylthiosemicarbazone); L3, diacetyl-bis(N(4)-methylthiosemicarbazone); L4, diacetyl-bis(N(4)-ethylthiosemicarbazone)} were synthesised and screened for activity against the human immunodeficiency virus (HIV). Complexes 1-4 were characterised using (1)H-NMR and IR spectroscopy; and their purity established by micronanalysis. Complex 3 inhibited viral infection of TZM-bl cells by 98% (IC(50)=6.8±0.6μM) at a non toxic concentration of 12.5μM while complex 4 inhibited infection of these cells by 72 and 98% (IC(50)=5.3±0.4μM) at concentrations of 6.25 and 12.5μM respectively. The mechanism of inhibition of infection in TZM-bl cells is presumably as a result of the cytostatic or anti-proliferative activity that was observed for complex 4 in real time cell electronic sensing (RT-CES) and carboxyflourescein succinimidyl ester (CFSE) analysis. Treatment of T lymphocytes from HIV infected individuals with 4 decreased CD4+ T cell expression (p=0.0049) as demonstrated by multi-parametric flow cytometry without suppressing cytokine production. None of the ligands (L1-L4) demonstrated anti-viral activity, supporting the importance of metal (gold) complexation in these potential drugs. Complexes 3 and 4 were shown to have ideal lipophilicity values that were similar when shake flask (0.97±0.5 and 2.42±0.6) and in silico prediction (0.8 and 1.5) methods were compared. The activity and drug-like properties of complexes 3 and 4 suggests that these novel metal-based compounds could be combined with virus inhibitory drugs to work as cytostatic agents in the emerging class of anti-HIV drugs known as virostatics.
Collapse
Affiliation(s)
- Pascaline N Fonteh
- Department of Biochemistry, University of Pretoria, Hatfield Campus, Pretoria 0002, South Africa
| | | | | |
Collapse
|
10
|
Uglietti A, Maserati R. Antiviral hyperactivation-limiting therapeutics as a novel class for the treatment of HIV/AIDS: focus on VS411. Expert Opin Investig Drugs 2011; 20:559-65. [DOI: 10.1517/13543784.2011.565047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Alessia Uglietti
- Fondazione “IRCCS Policlinico San Matteo”, Infectious Disease Department, Piazzale Golgi 19, 27100 Pavia, Italy
| | - Renato Maserati
- Fondazione “IRCCS Policlinico San Matteo”, Infectious Disease Department, Piazzale Golgi 19, 27100 Pavia, Italy
| |
Collapse
|
11
|
De Forni D, Stevens MR, Lori F. Strategies to improve efficacy and safety of a novel class of antiviral hyper-activation-limiting therapeutic agents: the VS411 model in [corrected] HIV/AIDS. Br J Pharmacol 2010; 161:830-43. [PMID: 20860662 DOI: 10.1111/j.1476-5381.2010.00940.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Antiviral hyper-activation-limiting therapeutic agents (AV-HALTs) are a novel experimental drug class designed to both decrease viral replication and down-regulate excessive immune system activation for the treatment of chronic infections, including human immunodeficiency virus (HIV)/acquired immunodeficiency syndrome. VS411, a first-in-class AV-HALT, is a single-dosage form combining didanosine (ddI, 400 mg), an antiviral (AV), and hydroxyurea (HU, 600 mg), a cytostatic agent, designed to provide a slow release of ddI to reduce its maximal plasma concentration (C(max)) to potentially reduce toxicity while maintaining total daily exposure (AUC) and the AV activity. EXPERIMENTAL APPROACH This was a pilot phase I, open-label, randomized, single-dose, four-way crossover trial to investigate the fasted and non-fasted residual variance of AUC, C(max) and the oral bioavailability of ddI and HU, co-formulated as VS411, and administered as two different fixed-dose combination formulations compared to commercially available ddI (Videx EC) and HU (Hydrea) when given simultaneously. KEY RESULTS Formulation VS411-2 had a favourable safety profile, displayed a clear trend for lower ddI C(max) (P= 0.0603) compared to Videx EC, and the 90% confidence intervals around the least square means ratio of C(max) did not include 100%. ddI AUC(∞) was not significantly decreased compared to Videx EC. HU pharmacokinetic parameters were essentially identical to Hydrea, although there was a decrease in HU exposure under fed versus fasted conditions. CONCLUSIONS AND IMPLICATIONS A phase IIa trial utilizing VS411-2 formulation has been fielded to identify the optimal doses of HU plus ddI as an AV-HALT for the treatment of HIV disease.
Collapse
|
12
|
Benito JM, López M, Lozano S, Ballesteros C, González-Lahoz J, Soriano V. Hydroxyurea exerts an anti-proliferative effect on T cells but has no direct impact on cellular activation. Clin Exp Immunol 2007; 149:171-7. [PMID: 17511775 PMCID: PMC1942020 DOI: 10.1111/j.1365-2249.2007.03412.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Hydroxyurea (HU) is a cytostatic drug which has been used as an anti-HIV agent due mainly to its synergistic activity when combined with certain anti-retrovirals. In addition, HU might have a beneficial effect on parameters involved in the pathogenesis of HIV infection, such as immune activation. To test this hypothesis, the effect of HU on T cell proliferation and T cell activation, as well as the potential association between these two phenomena, were examined in an in vitro model. HU exerted a dose-dependent anti-proliferative effect on T cells, and modulated the expression of different activation markers. In cells exposed to HU, expression of CD25 and CD38 diminished in a dose-dependent manner, whereas expression of CD69 increased. However, when the expression of these markers was examined separately on proliferating and non-proliferating lymphocytes, HU did not exert any significant effect. Thus, the effect of HU on T cell activation is not direct and seems to be mediated through its effect on T cell proliferation.
Collapse
Affiliation(s)
- J M Benito
- Department of Infectious Diseases, Hospital Carlos III, Madrid, Spain.
| | | | | | | | | | | |
Collapse
|
13
|
Oguariri RM, Brann TW, Imamichi T. Hydroxyurea and interleukin-6 synergistically reactivate HIV-1 replication in a latently infected promonocytic cell line via SP1/SP3 transcription factors. J Biol Chem 2006; 282:3594-604. [PMID: 17150965 DOI: 10.1074/jbc.m608150200] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The existence of viral latency limits the success of highly active antiretroviral therapy. With the therapeutic intention of reactivating latent virus to induce a cure, in this study we assessed the impact of cell synchronizers on HIV gene activation in latently infected U1 cells and investigated the molecular mechanisms responsible for such effect. Latently infected U1 cells were treated with 10 drugs including hydroxyurea (HU) and HIV-1 replication monitored using a p24 antigen capture assay. We found that HU was able to induce HIV-1 replication by 5-fold. HU has been used in the clinical treatment of HIV-1-infected patients in combination with didanosine; therefore, we investigated the impact of HU on HIV-1 activation in the presence of the proinflammatory cytokines, interleukin 6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha). IL-6 or TNF-alpha alone induced HIV replication by 18- and approximately 500-fold, respectively. Of interest, in the presence of HU, IL-6-mediated HIV-1 activation was enhanced by >90-fold, whereas TNF-alpha-mediated activation was inhibited by >30%. A reporter gene assay showed that HU and IL-6 synergized to activate HIV promoter activity via the Sp1 binding site. Electrophoretic mobility shift and supershift assays revealed increased binding of the Sp1 and Sp3 transcription factors to this region. Western blot analysis showed that HU and IL-6 co-stimulation resulted in increased levels of Sp1 and Sp3 proteins. In contrast, treatment with HU plus TNF-alpha down-regulated the expression of NF-kappaB. These findings suggest that Sp1/Sp3 is involved in controlling the HU/IL-6-induced reactivation of HIV-1 in latently infected cells.
Collapse
Affiliation(s)
- Raphael M Oguariri
- Laboratory of Human Retrovirology, Clinical Services Program, Science Applications International Corporation-Frederick Inc., NCI-Frederick, National Institutes of Health, Frederick, Maryland 21702, USA
| | | | | |
Collapse
|
14
|
Abacavir and Mycophenolic Acid, an Inhibitor of Inosine Monophosphate Dehydrogenase, Have Profound and Synergistic Anti-HIV Activity. ACTA ACUST UNITED AC 2006. [DOI: 10.1097/01.pas.0000213299.11649.101] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
15
|
Lori F, Foli A, Groff A, Lova L, Whitman L, Bakare N, Pollard RB, Lisziewicz J. Optimal suppression of HIV replication by low-dose hydroxyurea through the combination of antiviral and cytostatic ('virostatic') mechanisms. AIDS 2005; 19:1173-81. [PMID: 15990570 DOI: 10.1097/01.aids.0000176217.02743.d1] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The hydroxyurea-didanosine combination has been shown to limit immune activation (a major pathogenic component of HIV/AIDS) and suppress viral load by both antiviral and cytostatic ('virostatic') activities. Virostatics action represent a novel approach to attack HIV/AIDS from multiple directions; however, the use of these drugs is limited by the lack of understanding of their dose-dependent mechanism of action and by fear of pancreatic toxicity, even though a large review of ACTG studies has shown that hydroxyurea does not increase the incidence of pancreatitis. METHODS In vitro cytostatic and cytotoxic activity, inhibition of viral replication and immune activation by pharmacologically attainable plasma concentrations of hydroxyurea (10-100 micromol/l) and didanosine (1-5 micromol/l) were analyzed by cell proliferation, viability, apoptosis and infection assays using peripheral blood mononuclear cells. In vivo, 600, 900 and 1200 mg daily doses of hydroxyurea in combination with standard doses of didanosine and stavudine were studied in 115 randomized chronically infected patients. RESULTS A cytostatic low (10 micromol/l) concentration of hydroxyurea inhibited cell proliferation and HIV replication in vitro. A gradual switch from cytostatic to cytotoxic effects was observed by increasing hydroxyurea concentration to 50-100 micromol/l, predicting that lower doses of hydroxyurea would be less toxic and more potent in vivo. The clinical results confirmed that 600 mg hydroxyurea was better tolerated, had fewer side effects and was more potent in suppressing HIV replication than the higher doses. CONCLUSIONS A bimodal, dose-dependent, cytostatic-cytotoxic switch is an immune-based mechanism explaining the apparent paradox that lowering the dose of hydroxyurea to 600 mg daily induces maximal antiviral suppression in HIV-infected patients.
Collapse
Affiliation(s)
- Franco Lori
- Research Institute for Genetic and Human Therapy, IRCCS Policlinico S. Matteo, Pavia, Italy and Washington, DC 2007, USA
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Paton NI, Aboulhab J. Hydroxychloroquine, hydroxyurea and didanosine as initial therapy for HIV-infected patients with low viral load: safety, efficacy and resistance profile after 144 weeks. HIV Med 2005; 6:13-20. [PMID: 15670247 DOI: 10.1111/j.1468-1293.2005.00259.x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
OBJECTIVES To evaluate the long-term safety and efficacy of the combination of hydroxychloroquine, hydroxyurea and didanosine. METHODS We recruited antiretroviral-naive patients with viral loads less than 100 000 HIV-1 RNA copies/mL and CD4 counts greater than 150 cells/microL. All patients received hydroxychloroquine (200 mg), hydroxyurea (500 mg) and didanosine (125-200 mg) twice daily. Clinical and laboratory safety assessments and measurements of viral load and CD4 count were made at regular intervals, and genotypic resistance testing was performed on samples with detectable viral load at 48, 96 and 144 weeks. RESULTS Fourteen of the 17 patients who commenced therapy remained on treatment at 144 weeks. Treatment was well tolerated but caused neutropenia, usually mild and transient, in 12 patients (71%). Mean viral load was reduced by 1.6 log(10) copies/mL below baseline (P<0.001), eight patients (47%) had undetectable viral load (<400 copies/mL), and two patients (12%) had detectable viral load but no detectable resistance mutations at week 144. Four patients (24%) had detectable viral load together with major resistance mutations (three with both 74 V and 184 V, and one with both 62 V and 65R) at week 144, but still had viral load suppression below baseline. Mean CD4 count was increased by 106 cells/microL above baseline (P=0.07) at week 144. CONCLUSIONS This novel and well-tolerated combination controls viral replication during long-term follow up, with development of few resistance mutations. With careful monitoring it may be a useful strategy for delaying highly active antiretroviral therapy (HAART) and associated toxicity in selected patients with low initial viral loads.
Collapse
Affiliation(s)
- N I Paton
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore.
| | | |
Collapse
|
17
|
Lori F, Pollard RB, Whitman L, Bakare N, Blick G, Shalit P, Foli A, Peterson D, Tennenberg A, Schrader S, Rashbaum B, Farthing C, Herman D, Norris D, Greiger P, Frank I, Groff A, Lova L, Asmuth D, Lisziewicz J. Lowering the dose of hydroxyurea minimizes toxicity and maximizes anti-HIV potency. AIDS Res Hum Retroviruses 2005; 21:263-72. [PMID: 15943568 DOI: 10.1089/aid.2005.21.263] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The goal of this study was to optimize the hydroxyurea dosage in HIV-infected patients, and to minimize the toxicity and maximize the antiviral efficacy of the hydroxyurea-didanosine combination. In a randomized, open-label study (RIGHT 702, a multicenter trial performed in private and institutional practices), three daily doses (600 microg, 800-900 microg, and 1200 microg) of hydroxyurea were administered in combination with didanosine and stavudine to 115 chronically HIV-infected patients, one-third antiretroviral drug naive, with viremia between 5000 and 200,000 copies/ml regardless of CD4+ cell count. The primary efficacy end point was the proportion of patients with plasma HIV-1 RNA levels below 400 copies/ml after 24 weeks of therapy. In the RIGHT 702 intent-to-treat population the lowest (600 mg) dose of hydroxyurea was better tolerated, associated with fewer adverse events, and more potent by all efficacy parameters, including the primary end point (76 versus 60% patients with viremia<400 copies/ml at week 24 for the 600-mg and 800- to 900-mg dose groups, respectively; p=0.027), the mean area under the curve (60.3 versus 65.8; p=0.016), and the mean log10 decrease (-1.95 versus -0.77; p=0.001). Patients receiving 600 mg of hydroxyurea daily also had the highest CD4+ cell count, CD4+/CD8+ cell ratio, and lowest CD8+ cell count and percentage (p=0.035). The RIGHT 702 trial provides an explanation for the increased toxicity and decreased efficacy of hydroxyurea when it was used at high dosage (1200 mg daily). At the optimal dosage of 600 mg daily, hydroxyurea, in combination with didanosine, deserves reevaluation for the long-term management of HIV/AIDS worldwide, because of its excellent resistance profile, durability, and affordability.
Collapse
Affiliation(s)
- Franco Lori
- Research Institute for Genetic and Human Therapy (RIGHT), IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Lova L, Groff A, Ravot E, Comolli G, Xu J, Whitman L, Lewis M, Foli A, Lisziewicz J, Lori F. Hydroxyurea exerts a cytostatic but not immunosuppressive effect on T lymphocytes. AIDS 2005; 19:137-44. [PMID: 15668538 DOI: 10.1097/00002030-200501280-00005] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To demonstrate that, despite a dose-dependent cytostatic effect, hydroxyurea (HU) does not have immunosuppressive effects. METHODS The effects of HU on T lymphocyte proliferation parameters, activation phenotype and cytokine production were examined in vitro after exposure to clinically relevant concentrations of HU (10, 50, and 100 micromol/l). The effects of HU in vivo on CD4 T cell counts, viral load, activation phenotype and virus-specific response were examined in 17 Rhesus macaques infected with SIV(mac251) and randomized into three groups: untreated controls; treated with (R)-9-(2-phosphonylmethoxypropyl)adenine (PMPA) and didanosine (ddI) only; and treated with PMPA, didanosine, and HU. RESULTS The in vitro inhibition of T lymphocyte proliferation confirmed the cytostatic effect of HU, with a linear dose-dependent effect; however, no relevant differences were found in the expression of activation markers between treated and untreated controls. Both T helper type 1 and type 2 cytokine production were enhanced by HU. Consistent with the in vitro results, a blunted increase of peripheral CD4 T cells was observed in vivo in the HU group, without relevant effects on the expression of activation markers, and SIV-specific T cell responses were not affected by HU. CONCLUSIONS Hyper-proliferation of T-lymphocytes is a major factor contributing to HIV pathogenesis. HU exerts a cytostatic effect on T lymphocytes, without altering their activation and apparently without having an immunosuppressive effect. The increase in cytokine production at the single cell level might compensate for the decrease in the percentage of activated CD4 T lymphocytes, without overall impairment of HIV-specific immune responses.
Collapse
Affiliation(s)
- Luca Lova
- Research Institute for Genetic and Human Therapy at IRCCS Policlinico S. Matteo, Pavia, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Yang QE. Human immunodeficiency virus reservoir might be actively eradicated as residual malignant cells by cytotoxic chemotherapy. Med Hypotheses 2004; 62:358-63. [PMID: 14975503 DOI: 10.1016/j.mehy.2003.10.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2003] [Accepted: 10/06/2003] [Indexed: 11/24/2022]
Abstract
A unique characteristic of human immunodeficiency virus (HIV) infection is that the virus must incorporate its cDNA into the host genomic DNA for replication. Once the virus gets into the host genome and becomes a part of the host genetic materials, elimination of the virus without killing the infected cells is virtually impossible. The use of highly active antiretroviral therapy (HAART) can result in a substantial decline in viremia. However, HAART does not eradicate HIV. The progressive HIV infection will unavoidably rebound after a cessation of the treatment. Searching for a new combination therapeutic strategy with cytotoxic agents that eliminate or significantly reduce the HIV reservoir is a potential way for better control of the disease. Theoretically, the HIV reservoir can be gradually eradicated by long-term use of certain antimetabolic cytotoxic drugs coupled with proper activation of latently infected cells, if viral replication is completely blocked by antiretroviral chemotherapy to protect uninfected, susceptible cells.
Collapse
Affiliation(s)
- Quan-en Yang
- SAIC-Frederick, Inc., Laboratory of Antiviral Drug Mechanisms, National Cancer Institute at Frederick, Frederick, MD 21702, USA.
| |
Collapse
|
20
|
Malhotra U, Bosch RJ, Wang R, Collier A, McElrath MJ. Effect of adjunct hydroxyurea on helper T cell immunity in HIV type 1-infected patients with virological suppression. AIDS Res Hum Retroviruses 2004; 20:807-12. [PMID: 15320983 DOI: 10.1089/0889222041725226] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Hydroxyurea (HU) has preferential activity in virus reservoirs not effectively targeted by current antiretroviral drug regimens, but concern for potential toxicity has precluded its routine use. The effect of adjunct HU on T cell proliferative responses and phenotypic markers was examined in a randomized study of 39 chronically HIV-1-infected patients with virological suppression on potent antiretroviral therapy. While patients in the HU arm showed modest declines in the median CD4(+) T cell counts (total, -151 cells/mm(3); naive, -91 cells/mm(3)), no significant differences were noted in the Candida, HIV-1 p24, and HIV-1 gp160 responses between the treatment arms following 24 weeks of therapy. In conclusion, although adjunct HU was associated with modest declines in the CD4(+) T cell counts, there was no significant adverse effect on helper T cell function. Further trials to address the role of HU in HIV-1 treatment may be appropriate after careful selection of HU dose and the adjunct drugs to avoid nonhematological toxicity.
Collapse
Affiliation(s)
- Uma Malhotra
- Program in Infectious Diseases, Clinical Research Division, Fred Hutchinson Cancer Research Center, Department of Medicine, University of Washington School of medicine, Seattle, Washington, USA.
| | | | | | | | | |
Collapse
|
21
|
López M, Benito JM, Lozano S, Barreiro P, Martínez P, González-Lahoz J, Soriano V. Enhanced HIV-specific immune responses in chronically HIV-infected patients receiving didanosine plus hydroxyurea. AIDS 2004; 18:1251-61. [PMID: 15362657 DOI: 10.1097/00002030-200406180-00003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND The role of hydroxyurea (HU) in the treatment of HIV infection remains controversial. HU potentiates didanosine (ddI) antiviral activity and might exert immunomodulatory effects. PATIENTS AND METHODS Immunologic parameters were examined in HIV-infected patients enrolled in a simplification trial in which ddI-HU was provided to subjects who had been on complete virus suppression under highly active antiretroviral therapy (HAART) for longer than 6 months. A total of 84 of these patients showed plasma viraemia repeatedly below 5000 HIV-RNA copies/ml, and were the main study population. A group of 22 patients who continued on HAART and another of 22 drug-naive HIV-positive individuals were taken as controls. RESULTS At 12 months, the levels of naive and memory T-cell subsets were similar in patients on ddI-HU and under HAART, whereas immune activation tended to be lower in the former group. The frequency of HIV-specific CD8+ T cells (CTL) directed against 125 peptides derived from Gag, Pol, Env, Nef and HIV regulatory proteins was similar among patients on ddI-HU and untreated controls, and significantly higher than in patients under HAART. This higher CTL response in patients on ddI-HU was seen even when considering only subjects with undetectable viral load. HIV-specific CD4+ T-cell responses were absent in almost all patients on HAART, whereas they were present in up to 19% of patients on ddI-HU. CONCLUSION Treatment with ddI-HU provides higher levels of HIV-specific CD8+ and CD4+ T-cell responses than standard triple drug regimens. Thus, hydroxyurea might exert a beneficial immunomodulatory effect in HIV infection.
Collapse
Affiliation(s)
- Mariola López
- Service of Infectious Diseases, Hospital Carlos III. Madrid, Spain
| | | | | | | | | | | | | |
Collapse
|
22
|
de Mendoza C, Paxinos E, Barreiro P, Camino N, Núñez M, Soriano V. Different viral rebound following discontinuation of antiretroviral therapy in cases of infection with viruses carrying L74V or thymidine-associated mutations. J Clin Microbiol 2004; 42:862-6. [PMID: 14766874 PMCID: PMC344444 DOI: 10.1128/jcm.42.2.862-866.2004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
A total of 76 patients discontinued treatment with didanosine plus hydroxyurea after 1 year of maintenance therapy. The greatest human immunodeficiency virus (HIV)-RNA rebounds were seen in 10 patients harboring an L74V mutation, and the presence of viruses with this mutation rapidly waned. In contrast, viral rebounds were significantly less pronounced (P < 0.01) in 12 subjects harboring thymidine-associated mutations; these mutations persisted in all instances. Thus, selection of an L74V mutation during didanosine therapy may compromise HIV replication in vivo.
Collapse
|
23
|
Blanckenberg DH, Wood R, Horban A, Beniowski M, Boron-Kaczmarska A, Trocha H, Halota W, Schmidt RE, Fatkenheuer G, Jessen H, Lange JMA. Evaluation of nevirapine and/or hydroxyurea with nucleoside reverse transcriptase inhibitors in treatment-naive HIV-1-infected subjects. AIDS 2004; 18:631-40. [PMID: 15090768 DOI: 10.1097/00002030-200403050-00007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To examine the effect of adding nevirapine (NVP) and/or hydroxyurea (HU) to a triple nucleoside analogue reverse transcriptase inhibitor (NRTI) regimen in terms of efficacy and tolerability. METHODS : HIV-1-infected, treatment-naive adults were randomized, using a factorial design, to add NVP and/or HU to the triple NRTI backbone of zidovudine plus lamivudine plus abacavir. Primary endpoint was treatment failure, defined as having plasma HIV RNA levels > 50 copies/ml after week 24, or discontinuation of randomized treatment. Follow-up was 72 weeks. RESULTS For the 229 subjects, median plasma HIV-1 RNA was 4.61 log10 copies/ml and median CD4 cell count was 269 x 10 cells/l. NVP users reached plasma HIV-1 RNA < 50 copies/ml more rapidly than subjects using no NVP (log-rank test; P = 0.011). In the as-treated analysis, 21.6% of subjects using NVP versus 48.8% using no NVP reached the primary endpoint (P = 0.013). In the intent-to-treat analysis, 83.3% of subjects using HU versus 73.0% using no HU experienced treatment failure (P = 0.060), while no difference was observed in the as-treated analysis (34.5 versus 36.7%). Differences in the intent-to-treat analysis were accounted for by toxicity: 52.6% of subjects using HU experienced toxicity leading to discontinuation of randomized treatment versus 28.7% of subjects using no HU. CONCLUSION The use of NVP in addition to a triple NRTI regimen improved both short- and long-term antiretroviral efficacy. The use of HU significantly contributed to treatment failure because of toxicity.
Collapse
Affiliation(s)
- Daniel H Blanckenberg
- International Antiviral Therapy Evaluation Center, Departmernt of Human Retrovirology, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Foli A, Maserati R, Barasolo G, Castelli F, Tomasoni L, Migliorino M, Maggiolo F, Pan A, Paolucci S, Scudeller L, Tinelli C, D'Aquila R, Lisziewicz J, Lori F. Strategies to Decrease Viral Load Rebound, and Prevent Loss of Cd4 and Onset of Resistance during Structured Treatment Interruptions. Antivir Ther 2004. [DOI: 10.1177/135965350400900114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background Toxicity and other drug adherence-related factors have contributed to decreased compliance to antiretroviral regimens amongst HIV-infected patients. Irregular therapy disruption causes loss of CD4 T cells, onset of drug resistance and rapid rebound of plasma viral load (VL). However, an appropriate choice of drugs and properly scheduled structured treatment interruptions (STIs) may limit VL rebound, maintain CD4 counts and minimize resistance. Methods We conducted a clinical study of STIs, RIGHT 901, involving 60 drug-naive patients with chronic HIV infection (CD4 >300, VL >10000) randomized to receive didanosine-stavudine-indinavir (IDV group) or didanosine-stavudine-hydroxyurea (HU group), for 12 weeks. Subsequently, all patients were randomized again to start STI (short induction) or to continue the therapy for an additional 24 weeks before starting STI (long induction). Both groups underwent four STI cycles and then stopped therapy as long as viraemia remained below 10000 copies/ml before reinitiating another four cycles of STI. Results During continuous therapy VLs were suppressed at similar rates in both the HU and IDV groups, while a blunted CD4 count was documented in the HU group. Following the first stop median VL rebounded close to baseline values in both groups, however, during the following STI median VL rebound decreased in the HU group, while in the IDV group VL continued to rebound to values close to baseline, and the difference between the two groups was statistically significant. Moreover, patients treated with HU had a constant and stable CD4 increase during STI, whereas CD4 counts fluctuated in the IDV group, with sharp falls during treatment interruptions and partial CD4 recovery following treatment restart. Even in the presence of IDV resistance predisposing mutations at baseline, no genotypic change in the protease sequence was observed during STI. A relevant mutation in the reverse transcriptase sequence (K70R) emerged in one patient interrupting treatment after 36 weeks of continuous therapy and in one patient after four STI cycles. Side effects (no major events) were similar among groups. Conclusions An appropriate choice of STI schedule and regimens containing drugs less prone to resistance and/or able to prevent CD4 fluctuation may contribute to optimizing STI for chronically infected patients with respect to limiting viral rebound, improving CD4 counts and maintaining a resistance profile comparable to continuous highly active antiretroviral therapy.
Collapse
Affiliation(s)
- Andrea Foli
- RIGHT at IRCCS Policlinico S Matteo, Pavia, Italy and Washington, DC, USA
| | - Renato Maserati
- Infectious Diseases, IRCCS Policlinico S Matteo, Pavia, Italy
| | | | | | | | | | | | | | | | | | - Carmine Tinelli
- Direzione Scientifica, IRCCS Policlinico S Matteo, Pavia, Italy
| | - Richard D'Aquila
- Division of Infectious Diseases, Vanderbilt University, Nashville, Tenn., USA
| | | | - Franco Lori
- RIGHT at IRCCS Policlinico S Matteo, Pavia, Italy and Washington, DC, USA
| |
Collapse
|
25
|
Lisziewicz J, Foli A, Wainberg M, Lori F. Hydroxyurea in the treatment of HIV infection: clinical efficacy and safety concerns. Drug Saf 2003; 26:605-24. [PMID: 12814330 DOI: 10.2165/00002018-200326090-00002] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Data from basic science and clinical studies suggest that hydroxyurea (hydroxycarbamide)-based regimens are effective treatment options for patients with HIV at various stages of disease. In vitro studies of HIV-infected lymphocytes have shown that hydroxyurea: (i) inhibits viral DNA synthesis; (ii) synergistically interacts with nucleoside reverse transcriptase inhibitors (NRTI); and (iii) increases the antiviral activity of didanosine. Clinical studies have confirmed that hydroxyurea in combination with didanosine produces potent and sustained viral suppression in patients with HIV infection. However, some concerns have been recently raised on the use of hydroxyurea in association with NRTI. Hydroxyurea can cause myelosuppression, skin toxicities, mild gastrointestinal toxicity, and abnormalities of renal and liver functions. In addition, hydroxyurea may accentuate the toxic effects of nucleoside analogues. In fact, some clinical data seem to indicate an increased risk of pancreatitis and neuropathy when hydroxyurea is combined with didanosine and stavudine. Since hydroxyurea-related toxicity is dose dependent, a systematic study of hydroxyurea optimal dosage and schedule was initiated to monitor patients for possible nucleoside toxicity. In the Research Institute for Genetic and Human Therapy (RIGHT) 702 study it was shown that a low, well-tolerated hydroxyurea dose (600 mg daily) achieved better antiretroviral activity than higher doses, together with better CD4+ cell count increase and fewer adverse effects. In this paper the effects of hydroxyurea as salvage therapy for heavily pretreated patients with advanced HIV disease are presented. These studies have shown that some patients with extensive pretreatment experience and advanced disease can respond substantially to the addition of hydroxyurea. The addition of hydroxyurea to didanosine does not prevent the emergence of resistance to didanosine; nonetheless, the efficacy of this therapeutic regimen may not be attenuated by the presence of didanosine-resistant HIV mutants. Since CD4 T lymphocyte activation is essential for virus replication and CD8 T lymphocyte activation may contribute to pathogenesis, the combination of hydroxyurea with other drugs may lead to the inhibition of HIV, by blocking the 'cell activation-virus production-pathogenesis' cycle. Clinical data indicate that hydroxyurea may play a role in attenuation of viral rebound and immune reconstitution by decreasing CD4 T cell proliferation, as well as preventing the exhaustion of CD8 T cell populations that may result from excessive activation during HIV infection. While the combination of hydroxyurea with didanosine has provided hope, future studies including those that evaluate optimal dosing and long-term toxicity are needed to define the role for this agent in the treatment of HIV infection.
Collapse
|
26
|
Dogruel M, Gibbs JE, Thomas SA. Hydroxyurea transport across the blood-brain and blood-cerebrospinal fluid barriers of the guinea-pig. J Neurochem 2003; 87:76-84. [PMID: 12969254 DOI: 10.1046/j.1471-4159.2003.01968.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Hydroxyurea is used in the treatment of HIV infection in combination with nucleoside analogues, 2'3'-didehydro-3'deoxythymidine (D4T), 2'3'-dideoxyinosine or abacavir. It is distributed into human CSF and is transported from the CSF to sub-ependymal brain sites, but its movement into the brain directly from the blood has not been studied. This study addressed this by a brain perfusion technique in anaesthetized guinea-pigs. The carotid arteries were perfused with an artificial plasma containing [14C]hydroxyurea (1.6 microm) and a vascular marker, [3H]mannitol (4.6 nm). Brain uptake of [14C]hydroxyurea (8.0 +/- 0.9%) was greater than [3H]mannitol (2.4 +/- 0.2%; 20-min perfusion, n = 8). CSF uptake of [14C]hydroxyurea (5.6 +/- 1.5%) was also greater than [3H]mannitol (0.9 +/- 0.3%; n = 4). Brain uptake of [14C]hydroxyurea was increased by 200 microm hydroxyurea, 90 microm D4T, 350 microm probenecid, 25 microm digoxin, but not by 120 microm hydroxyurea, 16.5-50 microm D4T, 90 microm 2'3'-dideoxyinosine or 90 microm abacavir. [14C]Hydroxyurea distribution to the CSF, choroid plexus and pituitary gland remained unaffected by all these drugs. The metabolic half-life of hydroxyurea was > 15 h in brain and plasma. Results indicate that intact hydroxyurea can cross the brain barriers, but is removed from the brain by probenecid- and digoxin-sensitive transport mechanisms at the blood-brain barrier, which are also affected by D4T. These sensitivities implicate an organic anion transporter (probably organic anion transporting polypeptide 2) and possibly p-glycoprotein in the brain distribution of hydroxyurea and D4T.
Collapse
Affiliation(s)
- M Dogruel
- Centre for Neuroscience Research, Guy's, King's and St. Thomas School of Biomedical Science, King's College London, London, UK
| | | | | |
Collapse
|
27
|
Cooper DA. Update on didanosine. JOURNAL OF THE INTERNATIONAL ASSOCIATION OF PHYSICIANS IN AIDS CARE (CHICAGO, ILL. : 2002) 2003; 1:15-25. [PMID: 12942665 DOI: 10.1177/154510970200100105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Didanosine (ddl) has been a cornerstone of HIV management since it was made available in October 1991. Didanosine was originally introduced as an alternative to zidovudine (ZDV) for patients who were intolerant of ZDV or experienced disease progression during ZDV monotherapy. Didanosine is now used extensively as an integral component of multidrug combination regimens in both adults and children with HIV infection, and is now available for once-daily administration in the United States, Canada, and Europe. The recently approved Videx EC is an enteric-coated didanosine capsule dosed as one capsule, once daily. This paper provides an update of recently published studies on the use of ddl in combination anti-HIV therapy. In particular, these studies examine the rationale for the use of ddl as first-line anti-HIV therapy, and describe newer findings concerning its long-term efficacy, side effects, compliance, resistance, and once-daily use. The increased survival of HIV-infected patients is largely attributed to the introduction of the triple combination drug therapy but is probably also due to the long-term clinical efficacy of ddl.
Collapse
Affiliation(s)
- David A Cooper
- National Centre in HIV Epidemiology and Clinical Research, University of New South Wales, Sydney, Australia.
| |
Collapse
|
28
|
de Maat MMR, Ekhart GC, Huitema ADR, Koks CHW, Mulder JW, Beijnen JH. Drug interactions between antiretroviral drugs and comedicated agents. Clin Pharmacokinet 2003; 42:223-82. [PMID: 12603174 DOI: 10.2165/00003088-200342030-00002] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
HIV-infected individuals usually receive a wide variety of drugs in addition to their antiretroviral drug regimen. Since both non-nucleoside reverse transcriptase inhibitors and protease inhibitors are extensively metabolised by the cytochrome P450 system, there is a considerable potential for pharmacokinetic drug interactions when they are administered concomitantly with other drugs metabolised via the same pathway. In addition, protease inhibitors are substrates as well as inhibitors of the drug transporter P-glycoprotein, which also can result in pharmacokinetic drug interactions. The nucleoside reverse transcriptase inhibitors are predominantly excreted by the renal system and may also give rise to interactions. This review will discuss the pharmacokinetics of the different classes of antiretroviral drugs and the mechanisms by which drug interactions can occur. Furthermore, a literature overview of drug interactions is given, including the following items when available: coadministered agent and dosage, type of study that is performed to study the drug interaction, the subjects involved and, if specified, the type of subjects (healthy volunteers, HIV-infected individuals, sex), antiretroviral drug(s) and dosage, interaction mechanism, the effect and if possible the magnitude of interaction, comments, advice on what to do when the interaction occurs or how to avoid it, and references. This discussion of the different mechanisms of drug interactions, and the accompanying overview of data, will assist in providing optimal care to HIV-infected patients.
Collapse
Affiliation(s)
- Monique M R de Maat
- Department of Pharmacy and Pharmacology, Slotervaart Hospital, Amsterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
29
|
Mansky LM. Mutagenic outcome of combined antiviral drug treatment during human immunodeficiency virus type 1 replication. Virology 2003; 307:116-21. [PMID: 12667819 DOI: 10.1016/s0042-6822(02)00069-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The development of antiviral drug resistance is an important problem in the treatment of human immunodeficiency virus type 1 (HIV-1) infection. Potent antiretroviral therapy (ART) is currently used for treatment, and typically consists of at least two reverse transcriptase (RT) inhibitors. To assess the impact of combination therapy on HIV-1 mutagenesis, the mutagenic outcome of combined drug treatment was determined with several different RT drug combinations. Significant increases in HIV-1 mutant frequencies were observed with combinations of nucleoside RT inhibitors as well as in combinations where nucleoside inhibitors were used along with hydroxyurea, a drug known to deplete nucleotide pools in cells. This indicates that combinations of RT drugs can act together to further increase HIV-1 mutant frequencies, which could have important implications for virus population dynamics and could compromise drug therapy regimens.
Collapse
Affiliation(s)
- Louis M Mansky
- Department of Molecular Virology, Immunology, and Medical Genetics, 2078 Graves Hall, 333 West 10th Avenue, Ohio State University Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
30
|
Mansky LM, Le Rouzic E, Benichou S, Gajary LC. Influence of reverse transcriptase variants, drugs, and Vpr on human immunodeficiency virus type 1 mutant frequencies. J Virol 2003; 77:2071-80. [PMID: 12525642 PMCID: PMC140916 DOI: 10.1128/jvi.77.3.2071-2080.2003] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The evolution of drug resistance is a major complication of human immunodeficiency virus type 1 (HIV-1) chemotherapy. HIV-1 reverse transcriptase (RT) is a major target of antiretroviral therapy and ultimately the target of drug resistance mutations. Previous studies have indicated that drug-resistant HIV-1 RTs can alter HIV-1 mutant frequencies. In this study, we have tested a panel of HIV-1 RT variants for their ability to influence virus mutant frequencies. The RT variants tested included drug-resistant RT variants as well as other variants analyzed in enzyme fidelity studies with the lacZalpha gene as a mutation target and/or implicated as being important for enzyme fidelity by structural studies. Combinations of mutations that alone had a statistically significant influence on virus mutant frequencies resulted in different mutant frequency phenotypes. Furthermore, when virus replication occurred in the presence of drugs [e.g., 3'-azido-3'-deoxythymidine, (-)2/,3'-dideoxy-3'-thiacytidine, hydroxyurea, thymidine, or thioguanine] with selected RT variants, virus mutant frequencies increased. Similarly, Vpr variants deficient for binding to the uracil DNA glycosylase repair enzyme were observed to influence HIV-1 virus mutant frequencies when tested alone or in combination with RT variants. In summary, these observations indicate that HIV-1 mutant frequencies can significantly change by single amino acid substitutions in RT and that these effects can be altered by additional mutations in RT, by drugs, and/or by expression of Vpr variants. Such altered virus mutant frequencies could impact HIV-1 dynamics and evolution in small population sizes.
Collapse
Affiliation(s)
- Louis M Mansky
- Department of Molecular Virology, Immunology, and Medical Genetics, Center for Retrovirus Research, Comprehensive Cancer Center, Ohio State University Medical Center, Columbus, Ohio 43210, USA.
| | | | | | | |
Collapse
|
31
|
Abstract
The development of antiretroviral drug resistance is a major threat to the effective treatment of human immunodeficiency virus type 1 (HIV-1) infection. Drug treatment failure is associated with accumulation of drug resistance mutations and the evolution of drug resistance. Studies from microbial systems provide evidence for a correlation between drug resistance development and increased pathogen mutation rates. Recent studies with HIV-1 have shown that drugs targeted against reverse transcriptase (RT) as well as drug-resistant RT can increase HIV-1 mutation frequencies. Furthermore, combinations of drug and drug-resistant RT have been found to increase virus mutation frequencies in a multiplicative manner. The correlation of increased HIV-1 mutation rates with the evolution of antiretroviral drug resistance indicates that drug failure could increase the likelihood of further resistance evolving from subsequent drug regimens.
Collapse
Affiliation(s)
- Louis M Mansky
- Department of Molecular Virology, Immunology, and Medical Genetics, Center for Retrovirus Research, Ohio State University Medical Center, 2078 Graves Hall, 333 West 10th Avenue, Columbus 43210, USA.
| |
Collapse
|
32
|
Mayhew CN, Mampuru LJ, Chendil D, Ahmed MM, Phillips JD, Greenberg RN, Elford HL, Gallicchio VS. Suppression of retrovirus-induced immunodeficiency disease (murine AIDS) by trimidox and didox: novel ribonucleotide reductase inhibitors with less bone marrow toxicity than hydroxyurea. Antiviral Res 2002; 56:167-81. [PMID: 12367722 DOI: 10.1016/s0166-3542(02)00108-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Recently, the use of the ribonucleotide reductase (RR) inhibitor hydroxyurea (HU) in combination with nucleoside analogs has gained attention as a potential strategy for anti-HIV-1 therapy. However, appeal for the long-term use of HU in HIV-1 infection may be limited by its propensity to induce hematopoietic toxicity. We report a comparison of the efficacy and bone marrow toxicity of HU (400 and 200 mg/kg/day) with the novel RR inhibitors and free radical-scavenging compounds didox (DX; 3,4-dihydroxybenzohydroxamic acid; 350 mg/kg/day) and trimidox (TX; 3,4,5-trihydroxybenzamidoxime; 175 mg/kg/day) in the murine AIDS (LPBM5 MuLV) model of retrovirus infection. Infected mice received daily drug treatment for 8 weeks. Efficacy was determined by measuring drug effects on retroviral-induced disease progression (i.e. development of splenomegaly and hypergammaglobulinemia) and by evaluating splenic levels of proviral DNA. Bone marrow toxicity was evaluated by measuring peripheral blood indices (WBC, hematocrit and reticulocyte counts), femoral cellularity and by determining the numbers of hematopoietic progenitor cells (CFU-GM, BFU-E) per femur and spleen. Compared to infected controls receiving no drug treatment, disease progression was significantly suppressed by TX, DX and HU. However, HU was associated with mortality and induced significant hematopoietic toxicity in a time- and dose-dependent manner. Conversely, TX and DX effectively inhibited retrovirus-induced disease but did not induce hematopoietic toxicity. These results suggest that due to their reduced hematopoietic toxicity and ability to inhibit disease progression in murine AIDS, TX and DX may offer effective alternatives to HU therapy in HIV-1 infection.
Collapse
Affiliation(s)
- Christopher N Mayhew
- Department of Clinical Sciences, University of Kentucky Medical Center, CHS Building, 900 South Limestone Street, Lexington, KY 40536, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Mansky LM, Pearl DK, Gajary LC. Combination of drugs and drug-resistant reverse transcriptase results in a multiplicative increase of human immunodeficiency virus type 1 mutant frequencies. J Virol 2002; 76:9253-9. [PMID: 12186909 PMCID: PMC136424 DOI: 10.1128/jvi.76.18.9253-9259.2002] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Replication of drug-resistant human immunodeficiency virus type 1 (HIV-1) in the presence of drug can lead to the failure of antiretroviral drug treatment. Drug failure is associated with the accumulation of drug resistance mutations. Previous studies have shown that 3'-azido-3'-deoxythymidine (AZT), (-)2',3'-dideoxy-3'-thiacytidine (3TC), and AZT-resistant HIV-1 reverse transcriptase (RT) can increase the virus in vivo mutation rate. In this study, the combined effects of drug-resistant RT and antiretroviral drugs on the HIV-1 mutant frequency were determined. In most cases, a multiplicative effect was observed with AZT-resistant or AZT/3TC dually resistant RT and several drugs (i.e., AZT, 3TC, hydroxyurea, and thymidine) and led to increases in the odds of recovering virus mutants to over 20 times that of the HIV-1 mutant frequency in the absence of drug or drug-resistance mutations. This observation indicates that HIV-1 can mutate at a significantly higher rate when drug-resistant virus replicates in the presence of drug. These increased mutant frequencies could have important implications for HIV-1 population dynamics and drug therapy regimens.
Collapse
Affiliation(s)
- Louis M Mansky
- Department of Molecular Virology, Immunology, and Medical Genetics, Center for Retrovirus Research and Comprehensive Cancer Center, Ohio State University Medical Center, Columbus, Ohio 43210, USA.
| | | | | |
Collapse
|
34
|
Montaner JS, Harris M. Management of HIV-infected Patients with Multidrug-resistant Virus. Curr Infect Dis Rep 2002; 4:259-265. [PMID: 12015920 DOI: 10.1007/s11908-002-0089-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Heavily pretreated HIV-infected patients with multidrug-resistant virus remain a clinical challenge to the treating physician. While the goal of therapy in such patients is still controversial, sustained immunologic and clinical benefit have only been demonstrated with complete suppression of plasma viral load below detectable levels. Expert use of resistance testing may help in the selection of the salvage regimen, and monitoring of plasma drug levels may help optimize the potency and tolerability, especially of complex, multiple drug regimens where adherence remains a critical determinant of treatment outcome. The potential roles of newer agents, adjuvants, treatment interruptions, and immune-based therapies remain under investigation.
Collapse
Affiliation(s)
- Julio S.G. Montaner
- University of British Columbia/St. Paul's Hospital, 667-1081 Burrard Street, Vancouver, BC V6Z 1Y6, Canada.
| | | |
Collapse
|
35
|
Foli A, Benvenuto F, Piccinini G, Bareggi A, Cossarizza A, Lisziewicz J, Lori F. Direct analysis of mitochondrial toxicity of antiretroviral drugs. AIDS 2001; 15:1687-94. [PMID: 11546944 DOI: 10.1097/00002030-200109070-00012] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Mitochondrial toxicity is a serious side-effect of antiretroviral drugs, especially nucleoside reverse transcriptase inhibitors (NRTI). An in vitro assay to predict mitochondrial toxicity of in-use and developmental NRTI would be invaluable. To test the ability of a cytofluorimetric technique to predict the mitochondrial-dependent pancreatic and hepatic toxicity we used didanosine (ddI) alone or in combination with hydroxyurea (HU). METHODS The technique is based on the ability of the lipophilic cation JC-1 to enter selectively into mitochondria and change its colour as the membrane potential changes due to toxicity. Mitochondrial toxicity by HU and ddI was evaluated in pancreatic and hepatic human cell lines. The results were expressed as mitochondrial toxicity index (MTI), ranging from 0 to 100: the negative control was 0, and 100 indicating maximal toxicity. RESULTS Dose-dependent pancreatic toxicity of ddI was evident after 14 days of culture (MTI 34 +/- 4 at 100 microM, 10 +/- 4 at 10 microM, 2 +/- 3 at 1 microM ddI). HU alone was not toxic (MTI 7 +/- 10 at 100 microM, 2 +/- 2 at 50 microM and 2 +/- 4 at 10 microM HU); however, HU increased the toxicity of high, but not low, concentrations of ddI. For example, the MTI of 10 microM ddI plus 50 microM HU was 54 +/- 9. Negligible mitochondrial toxicity was observed in the hepatic cell line exposed to ddI alone or in combination with HU. CONCLUSIONS This in vitro assay might have in vivo relevance. First, ddI-related pancreatitis is dose dependent, and is reported more frequently than hepatic failure, consistent with our in vitro results. Second, patients who developed pancreatitis during randomized, controlled trials were treated with HU in combination with 400 mg ddI once daily (high peak concentration of ddI in the blood). In contrast, no pancreatitis was observed when HU was combined with 200 mg ddI twice daily (low peak concentration of ddI). These in vivo results are consistent with our in vitro observation that HU increases pancreatic cell toxicity in the presence of high concentrations of ddI. The in vitro assay described here might be used to predict the mitochondrial toxicity of other NRTI, alone or in combination.
Collapse
Affiliation(s)
- A Foli
- Research Institute for Genetic and Human Therapy, IRCCS Policlinico S. Matteo, Pavia, Italy
| | | | | | | | | | | | | |
Collapse
|
36
|
Balzarini J, Camarasa MJ, Pérez-Pérez MJ, San-Félix A, Velázquez S, Perno CF, De Clercq E, Anderson JN, Karlsson A. Exploitation of the low fidelity of human immunodeficiency virus type 1 (HIV-1) reverse transcriptase and the nucleotide composition bias in the HIV-1 genome to alter the drug resistance development of HIV. J Virol 2001; 75:5772-7. [PMID: 11390579 PMCID: PMC114293 DOI: 10.1128/jvi.75.13.5772-5777.2001] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The RNA genome of the lentivirus human immunodeficiency virus type 1 (HIV-1) is significantly richer in adenine nucleotides than the statistically equal distribution of the four different nucleotides that is expected. This compositional bias may be due to the guanine-to-adenine (G-->A) nucleotide hypermutation of the HIV genome, which has been explained by dCTP pool imbalances during reverse transcription. The adenine nucleotide bias together with the poor fidelity of HIV-1 reverse transcriptase markedly enhances the genetic variation of HIV and may be responsible for the rapid emergence of drug-resistant HIV-1 strains. We have now attempted to counteract the normal mutational pattern of HIV-1 in response to anti-HIV-1 drugs by altering the endogenous deoxynucleoside triphosphate pool ratios with antimetabolites in virus-infected cell cultures. We showed that administration of these antimetabolic compounds resulted in an altered drug resistance pattern due to the reversal of the predominant mutational flow of HIV (G-->A) to an adenine-to-guanine (A-->G) nucleotide pattern in the intact HIV-1-infected lymphocyte cultures. Forcing the virus to change its inherent nucleotide bias may lead to better control of viral drug resistance development.
Collapse
Affiliation(s)
- J Balzarini
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, Minderbroederstraat 10, B-3000 Leuven, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Weinberg A. In vitro hydroxyurea decreases Th1 cell-mediated immunity. CLINICAL AND DIAGNOSTIC LABORATORY IMMUNOLOGY 2001; 8:702-5. [PMID: 11427414 PMCID: PMC96130 DOI: 10.1128/cdli.8.4.702-705.2001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2000] [Accepted: 03/15/2001] [Indexed: 11/20/2022]
Abstract
Hydroxyurea (HU) is used in the treatment of hematologic disorders and is sometimes added to antiretroviral combination therapy to potentiate human immunodeficiency virus (HIV) suppression. However, HU has toxic effects on rapidly dividing cells, including the effectors of the immune response. To determine whether HU affects specific T-cell responses, we measured lymphocyte proliferation and cytokine production in response to microbial antigen and mitogen stimulation in the presence of added HU (10 to 1,000 microM). HU treatment of peripheral blood mononuclear cells obtained from HIV-infected patients and uninfected controls decreased lymphocyte proliferation and gamma interferon production compared with untreated cells. Interleukin-2 (IL-2) and IL-10 production was not affected by HU. The HU-mediated decrease of lymphocyte proliferation was similar in peripheral blood mononuclear cells from HIV-infected patients and from uninfected controls. The inhibitory effect of HU required continuous exposure to the drug and could be reverted by washing the drug out of the culture environment. These findings suggest that HU-containing therapeutic regimens might decrease Th1-cell-mediated immune responses in vivo.
Collapse
Affiliation(s)
- A Weinberg
- Department of Pediatrics, University of Colorado Health Sciences Center, Denver, Colorado 80262, USA.
| |
Collapse
|
38
|
Sereti I, Lane HC. Immunopathogenesis of human immunodeficiency virus: implications for immune-based therapies. Clin Infect Dis 2001; 32:1738-55. [PMID: 11360217 DOI: 10.1086/320758] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2000] [Revised: 01/12/2001] [Indexed: 11/03/2022] Open
Abstract
Human immunodeficiency virus (HIV) infection leads to a state of CD4 lymphopenia and generalized immune activation with subsequent development of opportunistic infections and neoplasms. The use of highly active antiretroviral treatment has dramatically improved the clinical outcome for HIV-infected patients, but the associated cost and toxicity and the eventual development of drug resistance have underscored the need for additional therapeutic strategies. Immune-based therapies, such as treatment with cytokines or immunosuppressants, adoptive immunotherapy, and therapeutic immunizations, are being intensely investigated as potential supplements to antiretroviral therapy. Although much data have been generated as a result of these efforts, to date there has been little evidence of the clinical efficacy of these strategies. Randomized clinical studies remain critical in evaluating the clinical significance and the role of immune-based therapies in the therapeutic armamentarium against HIV.
Collapse
Affiliation(s)
- I Sereti
- Cellular and Molecular Retrovirology Section, Laboratory of Immunoregulation, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | | |
Collapse
|
39
|
Longhurst HJ, Pinching AJ. Drug Points: pancreatitis associated with hydroxyurea in combination with didanosine. BMJ (CLINICAL RESEARCH ED.) 2001; 322:81. [PMID: 11154621 PMCID: PMC26596 DOI: 10.1136/bmj.322.7278.81] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
40
|
Colebunders R, Florence E, Ostyn B. Systematic review of combination antiretroviral therapy with didanosine plus hydroxyurea: a partial solution to Africa's HIV/AIDS problem? Int J Infect Dis 2001; 5:234; author reply 234-5. [PMID: 11962349 DOI: 10.1016/s1201-9712(01)90081-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
41
|
Long-Term Safety and Antiretroviral Activity of Hydroxyurea and Didanosine in HIV-Infected Patients. J Acquir Immune Defic Syndr 2000. [DOI: 10.1097/00126334-200012010-00006] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
42
|
Biron F, Ponceau B, Bouhour D, Boibieux A, Verrier B, Peyramond D. Long-term safety and antiretroviral activity of hydroxyurea and didanosine in HIV-infected patients. J Acquir Immune Defic Syndr 2000; 25:329-36. [PMID: 11114833 DOI: 10.1097/00042560-200012010-00006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Long-term safety, immunologic effects, and antiretroviral activity of hydroxyurea and didanosine were evaluated in this retrospective study. Some 65 HIV-1-infected patients (39 of whom were antiretroviral naive) were studied (mean baseline CD4 count, 362 cells/mm3; mean plasma HIV-1 RNA viral load, 4.8 log10 copies/ml). The mean treatment duration was 20 months. Overall tolerance was good: 15 patients interrupted treatment because of clinical or biologic side effects. Four patients experienced a category B event. Patients had a mean increase of 27 CD4 cell counts after 12 months, of 112 after 24 months and of 59 after 36 months. They had a mean 1. 03 log10 fall in HIV-1 RNA after 12 months, 1.59 log10 after 24 months, and 1.27 log10 after 36 months. After 12 months, 35% developed an HIV-1 RNA viral load <200 copies/ml, 53% after 24 months, and 36% after 36 months. Those whose viral load became undetectable after 12 months have significantly lower baseline RNA values (p =.03). Fourteen patients had a viral load <3.4 log10 copies/ml after 24 months of the double therapy. A prolonged viral load suppression can be achieved using a simple combination of two drugs that are inexpensive and well tolerated.
Collapse
Affiliation(s)
- F Biron
- Croix-Rousse Hospital Medical Centre/University, Lyon, France.
| | | | | | | | | | | |
Collapse
|
43
|
Laughon SK, Shinn LL, Nunley JR. Melanonychia and mucocutaneous hyperpigmentation due to hydroxyurea use in an HIV-infected patient. Int J Dermatol 2000; 39:928-31. [PMID: 11168664 DOI: 10.1046/j.1365-4362.2000.00988-2.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- S K Laughon
- Department of Dermatology, Medical College, Virginia Campus, Virginia Commonwealth University, Richmond, Virginia, USA
| | | | | |
Collapse
|
44
|
Heredia A, Davis C, Redfield R. Synergistic inhibition of HIV-1 in activated and resting peripheral blood mononuclear cells, monocyte-derived macrophages, and selected drug-resistant isolates with nucleoside analogues combined with a natural product, resveratrol. J Acquir Immune Defic Syndr 2000; 25:246-55. [PMID: 11115955 DOI: 10.1097/00126334-200011010-00006] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Resveratrol (trans-3,5,4;-trihydroxystilbene) is a phytoalexin present in grapes, wine, and certain plants, which has recently been reported to possess properties that may protect against atherosclerosis, certain cancers, and inflammation. We now report that resveratrol (RV) synergistically enhances the anti-HIV-1 activity of the nucleoside analogues zidovudine (AZT), zalcitabine (ddC), and didanosine (ddI). RV at 10 microM was not toxic to cells, and by itself reduced viral replication by 20% to 30%. In phytohemagglutinin (PHA)-activated peripheral blood mononuclear cells (PBMCs) infected with HTLV-IIIB, 10 microM RV reduced the 90 % inhibitory concentration (IC90) of AZT, ddC, and ddI by 3.5-, 5.5-, and 17.8-fold, respectively. Similar antiviral activity was demonstrated when ddI was combined with 5 or 10 mM RV in PBMCs infected with clinical isolates of HIV-1. The addition of RV resulted in a >10-fold augmentation of ddI-antiviral activity in infected monocyte-derived macrophages (MDMs). In a resting cell model of T lymphocytes which were infected with HTLV-IIIB, RV plus ddI in combination, but not individually, suppressed establishment of a productive viral infection. In addition, RV plus ddI markedly inhibited the replication of four ddI-resistant viral isolates, three of which presented mutations in the RT gene conferring RT-multidrug resistance. Finally, when compared with hydroxyurea (HU), both 100 mM HU and 10 mM RV showed similar enhancement of ddI-antiviral suppressive activity. However, RV was shown to have less of a cellular antiproliferative effect than HU.
Collapse
Affiliation(s)
- A Heredia
- Institute of Human Virology, University of Maryland Biotechnology Institute and University of Maryland Medicine, Baltimore, MD 21201, USA
| | | | | |
Collapse
|
45
|
Kline MW, Calles NR, Simon C, Schwarzwald H. Pilot study of hydroxyurea in human immunodeficiency virus-infected children receiving didanosine and/or stavudine. Pediatr Infect Dis J 2000; 19:1083-6. [PMID: 11099091 DOI: 10.1097/00006454-200011000-00011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To evaluate the safety and antiviral and immunologic effects of hydroxyurea given with didanosine (ddI) and/or stavudine (d4T) to symptomatic HIV-infected children. METHODS HIV-infected children with a history of long term nucleoside antiretroviral therapy were treated orally with hydroxyurea (initial dose, 10 to 20 mg/kg once daily; final dose, 30 mg/kg once daily), added to existing therapy that included ddI and/or d4T. RESULTS Sixteen children were enrolled (mean age, 6.7 years; range, 1.8 to 13.4 years). Antiretroviral therapy used with hydroxyurea included d4T/ddI (12), ddI (2), d4T (1) and d4T/lamivudine (1). Children received between 24 and 48 weeks of therapy, which was well-tolerated. Hydroxyurea was held temporarily during the first month of therapy in 4 cases because of neutropenia; all patients resumed hydroxyurea at full dosage without recurrence of neutropenia. No patient discontinued therapy permanently because of intolerance or toxicity. For the 13 children who completed 48 weeks of study treatment, the mean plasma HIV RNA concentration decreased from 4.6 log10 copies/ml at baseline to 4.2 log10 copies/ml at study Week 48 (P = 0.035, paired t test). Eight of these 13 children experienced a 0.5-log10 copies/ml or greater drop in HIV RNA concentration in the 48 weeks of study treatment. Appreciable changes in CD4+ lymphocyte percentage were not noted. CONCLUSIONS Hydroxyurea, added to existing therapy with ddI and/or d4T, was well-tolerated and safe in HIV-infected children. Evidence of antiviral activity was observed in some cases.
Collapse
Affiliation(s)
- M W Kline
- Department of Pediatrics, Baylor College of Medicne, Texas Children's Hospital, Houston 77030, USA.
| | | | | | | |
Collapse
|
46
|
Synergistic Inhibition of HIV-1 in Activated and Resting Peripheral Blood Mononuclear Cells, Monocyte-Derived Macrophages, and Selected Drug-Resistant Isolates With Nucleoside Analogues Combined With a Natural Product, Resveratrol. J Acquir Immune Defic Syndr 2000. [DOI: 10.1097/00042560-200011010-00006] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
47
|
Rutschmann OT, Vernazza PL, Bucher HC, Opravil M, Ledergerber B, Telenti A, Malinverni R, Bernasconi E, Fagard C, Leduc D, Perrin L, Hirschel B. Long-term hydroxyurea in combination with didanosine and stavudine for the treatment of HIV-1 infection. Swiss HIV Cohort Study. AIDS 2000; 14:2145-51. [PMID: 11061656 DOI: 10.1097/00002030-200009290-00011] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE AND METHODS In 1998 we reported on a randomized comparison between stavudine plus didanosine plus placebo versus stavudine plus didanosine plus hydroxyurea (HU), in patients with a CD4 count of 200-500 x 10(6) cells/l. After 3 months, the HU group had a higher proportion of patients with viral load < 200 x 10 cells/l. At the end of the 3 months blinded period, patients in the placebo group had the option to add HU if their viral load remained > 200 x 10(6) cells/l. We report results after 24 months. RESULTS Seventy-two patients were randomized to the HU arm, and a further 30 elected to add HU after 12 weeks. Twenty-four months after the start of the trial, only 25% of the 72 patients originally randomized to HU, and 20% of the 30 who added HU after week 12, were still taking it. The reasons for stopping HU were: lack of efficacy (45%), adverse events (37%) and patient or physician preference (18%). Side effects were more frequent in the didanosine/stavudine/HU group than in the didanosine/stavudine group: neuropathy (35 versus 15%, P< 0.02), fatigue (22 versus 7%, P< 0.01), and nausea or vomiting (26 versus 9%, P< 0.01). Of those who had discontinued HU, 73% were taking three drugs including a protease inhibitor. Patients who had started HU were compared with similar patients who had started protease inhibitors in the Swiss cohort. The probability of stopping HU was higher than the probability of stopping nelfinavir or indinavir, and similar to the probability of stopping ritonavir. CONCLUSION HU increased the antiviral effect of stavudine plus didanosine. However, side effects were more frequent, and after 24 months the majority of patients had switched to protease inhibitor regimens.
Collapse
|
48
|
Balzarini J. Effect of antimetabolite drugs of nucleotide metabolism on the anti-human immunodeficiency virus activity of nucleoside reverse transcriptase inhibitors. Pharmacol Ther 2000; 87:175-87. [PMID: 11007999 DOI: 10.1016/s0163-7258(00)00050-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A number of attempts are currently underway to combine antimetabolite drugs of nucleotide metabolism with a nucleoside reverse transcriptase inhibitor (NRTI) targeting human immunodeficiency virus (HIV) to improve the antiviral efficacy of the NRTIs and to better control HIV drug resistance. Hydroxyurea, a ribonucleotide reductase inhibitor, is currently combined with the NRTI didanosine (2',3'-dideoxyinosine) in clinical trials. However, other cellular target enzymes, including thymidylate synthase, inosinate dehydrogenase, cytidine-5'-triphosphate synthetase, and other enzymes from the de novo nucleotide biosynthesis pathway, can also be considered to potentiate the antiviral action of NRTIs. The underlying reasons for the potentiation of the antiviral activity of the NRTIs by antimetabolite drugs of nucleotide metabolism can be multiple. Decreased endogenous 2'-deoxynucleoside-5'-triphosphate (dNTP) pools result in a better competition of the NRTI (as its triphosphate derivative), with the dNTPs for the virus-encoded reverse transcriptase to be recognized as a substrate for the DNA polymerization reaction and subsequently to be incorporated into the growing viral DNA chain. Also, an increased metabolism (phosphorylation) of the NRTI by stimulatory enzyme feedback mechanisms may result in the production of higher levels of NRTI triphosphate. Thus, higher intracellular ratios of NRTI-triphosphate/dNTP created by well-defined combinations of NRTIs and antimetabolite drugs enable a more profound inhibitory effect of the NRTI against the reverse transcriptase (and thus, against the virus) and a better suppression of resistant (mutant) virus strains. A profound evaluation of this relatively new concept in the clinical setting will reveal whether this approach will establish a place in future treatment modalities of HIV infections.
Collapse
Affiliation(s)
- J Balzarini
- Rega Institute for Medical Research, K. U. Leuven, Minderbroedersstraat 10, B-3000, Leuven, Belgium.
| |
Collapse
|
49
|
Lori F, Lisziewicz J. Rationale for the use of hydroxyurea as an anti-human immunodeficiency virus drug. Clin Infect Dis 2000; 30 Suppl 2:S193-7. [PMID: 10860905 DOI: 10.1086/313851] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Hydroxyurea has been extensively used in medical practice, mainly for treating chronic myelogenous leukemia, sickle cell anemia, and other diseases. In light of its ability to inhibit DNA synthesis and to induce cell cycle arrest through inhibition of ribonucleotide reductase, the effects of hydroxyurea on replication of human immunodeficiency virus type 1 (HIV-1) have been investigated. In vitro hydroxyurea has been shown to block HIV-1 reverse transcription and/or replication in quiescent peripheral blood mononuclear cells (PBMC) and macrophages. Hydroxyurea was also found to be synergistic with the nucleoside reverse transcriptase inhibitor didanosine and to inhibit HIV-1 replication in activated PBMC; this inhibition may be due to a reduction in deoxynucleoside triphosphate pool sizes. Finally, hydroxyurea has been shown to sensitize didanosine-resistant mutants. Hydroxyurea may therefore be useful for limiting the spread of didanosine-resistant HIV-1 variants. The favorable toxicity profile of hydroxyurea and the lack of significant overlapping toxicities with some of the nucleoside reverse transcriptase inhibitors, as well as their distinct mechanisms of action, have provided further rationale for use of these agents in combination therapies.
Collapse
Affiliation(s)
- F Lori
- Research Institute for Genetic and Human Therapy, Washington, DC 20007, USA.
| | | |
Collapse
|
50
|
Zala C, Rouleau D, Montaner JS. Role of hydroxyurea in treatment of disease due to human immunodeficiency virus infection. Clin Infect Dis 2000; 30 Suppl 2:S143-50. [PMID: 10860899 DOI: 10.1086/313856] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
The potential role of hydroxyurea in the treatment of human immunodeficiency virus (HIV) infection was first supported by in vitro experiments that demonstrated control of viral production in activated and resting T cells. More recently, controlled clinical trials demonstrated that the addition of hydroxyurea to nucleoside-including regimens (chiefly of didanosine but also of stavudine and lamivudine) enhances their antiviral potency. It is believed that the cytostatic effect of hydroxyurea is at least partially responsible for its antiviral effect, through the down-modulation of cellular proliferation. Such an effect has also been credited for the blunted CD4 T cell responses that are characteristically observed when hydroxyurea is added to nucleoside-including regimens. The adjunctive antiviral effect of hydroxyurea-as well as its favorable dosing schedule, safety profile, and cost-makes it a very attractive addition to our therapeutic armamentarium. Further research is urgently needed to delineate the most appropriate use of this compound in the setting of HIV antiretroviral therapy.
Collapse
Affiliation(s)
- C Zala
- Fundación Huésped, 1202 Buenos Aires, Argentina. . ar
| | | | | |
Collapse
|