Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Impact of Chemotherapy for HIV-1 Related Lymphoma on Residual Viremia and Cellular HIV-1 DNA in Patients on Suppressive Antiretroviral Therapy

Abstract

The first cure of HIV-1 infection was achieved through complex, multimodal therapy including myeloablative chemotherapy, total body irradiation, anti-thymocyte globulin, and allogeneic stem cell transplantation with a CCR5 delta32 homozygous donor. The contributions of each component of this therapy to HIV-1 eradication are unclear. To assess the impact of cytotoxic chemotherapy alone on HIV-1 persistence, we longitudinally evaluated low-level plasma viremia and HIV-1 DNA in PBMC from patients in the ACTG A5001/ALLRT cohort on suppressive antiretroviral therapy (ART) who underwent chemotherapy for HIV-1 related lymphoma without interrupting ART. Plasma HIV-1 RNA, total HIV-1 DNA and 2-LTR circles (2-LTRs) in PBMC were measured using sensitive qPCR assays. In the 9 patients who received moderately intensive chemotherapy for HIV-1 related lymphoma with uninterrupted ART, low-level plasma HIV-1 RNA did not change significantly with chemotherapy: median HIV-1 RNA was 1 copy/mL (interquartile range: 1.0 to 20) pre-chemotherapy versus 4 copies/mL (interquartile range: 1.0 to 7.0) post-chemotherapy. HIV-1 DNA levels also did not change significantly, with median pre-chemotherapy HIV-1 DNA of 355 copies/106 CD4+ cells versus 228 copies/106 CD4+ cells post-chemotherapy. 2-LTRs were detectable in 2 of 9 patients pre-chemotherapy and in 3 of 9 patients post-chemotherapy. In summary, moderately intensive chemotherapy for HIV-1 related lymphoma in the context of continuous ART did not have a prolonged impact on HIV-1 persistence.

Clinical Trials Registration Unique Identifier:

NCT00001137

Introduction

Effective antiretroviral therapy (ART) reduces plasma HIV-1 RNA to levels that are undetectable by FDA-approved assays, but low-level viremia and HIV-1 DNA in PBMC persist [1], [2]. The persistence of replication-competent HIV-1 in long-lived memory CD4+ T cells despite prolonged ART administration is a major obstacle to curing HIV-1 infection [3][5]. Nevertheless, in one HIV-infected individual, allogeneic hematopoietic stem cell transplantation (ASCT) with a CCR5 delta32/delta32 donor resulted in the first definitive cure of HIV-1 infection [6]. This cure has generated enthusiasm for further investigation of potentially curative interventions for HIV-1, including allogeneic stem cell transplantation [7], [8] and autologous transplantation with genetically modified CD4+ T cells [9] or stem cells [10], [11]. Along these lines, the National Heart, Lung and Blood Institute (NHLBI) recently identified the possible role of hematopoietic stem cells in curative approaches for HIV-1 infection as an essential question that needs to be addressed [12].

Although there is considerable interest in stem cell-mediated interventions to achieve a cure of HIV-1, the question remains as to which components of the Berlin patient's cancer therapy were necessary to achieve a cure. Components of ASCT that may have contributed to the eradication of HIV-1 reservoirs include chemotherapy, total body irradiation, immunosuppressive drugs, allogeneic transplantation with CCR5 delta32/delta32 donor cells and graft versus host disease. We have shown previously that myeloablative chemotherapy followed by autologous hematopoietic stem cell transplantation is not sufficient to eliminate low-level HIV-1 RNA in plasma or HIV-1 DNA in PBMC in patients on ART with <50 cps/mL of HIV-1 RNA in plasma, but changes from before myeloablative therapy to after autologous transplant were not compared in this prior study [13], thus the impact chemotherapy alone is undefined. Intensive chemotherapy is known to cause significant depletion of circulating CD4+ T cells [14][16], which could reduce levels of plasma viremia or HIV-1 DNA in PBMC in the context of uninterrupted ART by killing HIV-infected cells. To investigate this possibility, we measured HIV-1 levels in plasma and PBMC samples, before and after chemotherapy, in 9 patients who underwent moderately intensive chemotherapy for HIV-1 related lymphoma and who remained on continuous ART throughout the sampling period.

Methods

NWCS 334 was a retrospective study of HIV-1-infected patients in the ACTG A5001: AIDS Clinical Trials Group Longitudinal Linked Randomized Trials (ALLRT) cohort who received moderately intensive chemotherapy for the treatment of HIV-1 related lymphoma, who continued on ART pre- and post-chemotherapy, and who maintained suppressed plasma HIV-1 RNA<50 copies/mL (Roche Amplicor HIV Monitor assay versions 1.0/1.5; Branchburg, NJ). The ALLRT parent study is registered at ClinicalTrials.gov under the unique identifier NCT00001137, and the rationale, design, and baseline characteristics of the overall cohort have been previously described [17]. The University of Pittsburgh Institutional Review Board approved the parent study (ALLRT), which allowed participants to contribute samples for future use in ACTG-approved research. Patients gave written informed consent for the information to be obtained from their clinic records as part of the ALLRT study, and this information was kept confidential at each ALLRT site. Patient information was anonymized and de-identified prior to analysis.

Stored plasma samples were evaluated for HIV-1 RNA using two-step real-time quantitative PCR assays with two different primer/probe sets targeting HIV-1 gag or integrase sequences using previously described assay conditions with single-copy sensitivity (limit of detection<1 copy/mL of plasma) [13], [18]. Levels of total HIV-1 DNA (limit of quantification = 5 copies/sample) and 2-long terminal repeat circles (2-LTRs; limit of quantification = 7.5 copies/sample) in PBMC were assayed as described previously [13] and were run in parallel with positive and negative controls from the Virology Quality Assurance Laboratory (Rush University). HIV-1 DNA quantitative PCR (qPCR) data were normalized per 106 CD4+ T cells using qPCR for the CCR5 gene [19] and the percent CD4+ T cells. The percent CD4+ T cells was available from the A5001/ALLRT database (N = 8) or was determined by standard flow cytometry in the Pitt Virology Support Laboratory (N = 1).

Statistical analysis using McNemar's Test was applied to determine if the proportion of patients with plasma viremia at undetectable levels pre-chemotherapy was significantly different from the proportion of patients with undetectable levels post-chemotherapy. A non-parametric sign test was used to determine if there was a significant difference between CD4+ and CD8+ cell counts pre- and post-chemotherapy, and between HIV-1 DNA copies per 106 CD4+ T cells pre- and post-chemotherapy.

Results

A total of 40 patients in the A5001/ALLRT cohort were diagnosed with HIV-1 related lymphoma, 18 had plasma HIV-1 RNA<50 cps/mL pre- and post-chemotherapy with uninterrupted ART, and 10 of these 18 had plasma and PBMC samples available pre- and post-chemotherapy for further analysis. To confirm the efficiency of qPCR amplification for HIV-1 RNA, plasma samples from prior to the initiation of ART in these 10 patients were tested and results were compared to the FDA-approved Roche Amplicor assay. HIV-1 RNA in pre-ART samples from 9 of 10 subjects amplified efficiently by qPCR, and longitudinal samples from these 9 patients were studied. The relevant clinic characteristics of the study patients are shown in Table 1. All patients were males diagnosed with HIV-1 related Hodgkin's (HL; N = 3) or Non-Hodgkin's lymphoma (NHL; N = 6).

thumbnail
Table 1. Characteristics of the 9 patients studied from the ALLRT cohort.

https://doi.org/10.1371/journal.pone.0092118.t001

All 9 patients received moderately intensive chemotherapy for lymphoma consisting of doxorubicin, bleomycin, vinblastine and dacarbazine (ABVD) in 4 patients, cyclophosphamide, doxorubicin, vincristine, prednisone (CHOP) in 3 patients, and CHOP with rituximab in 2 patients (Table 1). The number of days between pre- and post-chemotherapy sampling varied, with a median of 285 days between samples (interquartile range: 225 to 685 days). Median CD4+ count pre-chemotherapy was 296 cells/μL (median %CD4 = 20.0) and was 315 cells/μL (median %CD4 = 20.0) post-chemotherapy; median CD8+ count pre-chemotherapy was 803 cells/μL (median %CD8 = 50.0%) and 693 cells/μL (median %CD8 = 49.0%) post-chemotherapy. Differences in the CD4+ and CD8+ cell counts between pre- and post-chemotherapy time points were not statistically significant.

All subjects received ART throughout their chemotherapy and post-chemotherapy follow-up. Low-level plasma HIV-1 RNA (Figure 1A), as determined by qPCR with single-copy sensitivity [13], did not show a consistent pattern of change from pre- to post-chemotherapy time points. Plasma viremia decreased in 5 patients (median decrease = 8 copies/mL), increased in 3 (median increase = 3 copies/mL), and remained below the LOD in one patient (Figure 1A). The median HIV-1 plasma RNA pre-chemotherapy was 1 copy/mL, and the median post-chemotherapy was 4 copies/mL. A total of 4 patients had undetectable plasma HIV-1 RNA pre-chemotherapy and 3 had undetectable plasma HIV-1 RNA post-chemotherapy. There was no significant difference in the proportion of patients with undetectable plasma HIV-1 RNA before versus after chemotherapy (p = 0.6, McNemar's test).

thumbnail
Figure 1. Quantification of HIV-1 from plasma and PBMC pre- and post-chemotherapy.

A) Plasma HIV-1 RNA decreased for 5 patients, increased for 3 patients, and was below the limit of detection at both time points for 1 patient. Median viral load pre-chemotherapy was 1 copy/mL versus 4 copies/mL post-chemotherapy. Open symbols indicate undetectable samples. B) Total HIV-1 DNA levels per 106 CD4+ cells decreased in 4 patients and increased in 5 patients. Median HIV-1 DNA levels per 106 CD4+ cells decreased from 355 to 228 copies per 106 CD4+ cells from pre- to post-chemotherapy. C) 2-LTRs were detectable pre-chemotherapy in 2 of 9 patients versus detectable in 3 of 9 post-chemotherapy. Patients with undetectable 2-LTRs at both time points are not shown. Open symbols indicate undetectable samples.

https://doi.org/10.1371/journal.pone.0092118.g001

Levels of total HIV-1 DNA and 2-LTRs in PBMC (Figures 1B and 1C) were evaluated for changes between pre- and post-chemotherapy time points. Total HIV-1 DNA was detectable in all 9 patients pre- and post-chemotherapy and showed no consistent pattern of change. The median total HIV-1 DNA level pre-chemotherapy was 355 (interquartile range: 70.0 to 469) copies/106 CD4+ T cells versus 228 (interquartile range: 106 to 452) copies/106 CD4+ cells post-chemotherapy (p = 1.0, sign test). Total HIV-1 DNA levels per 106 CD4+ cells decreased in 4 patients and increased in 5 patients between pre- and post-chemotherapy time points. 2-LTRs were detectable in only 4 of 9 subjects at either time point, with 2-LTRs increasing in 1 patient post-chemotherapy, becoming detectable (from below the limit of quantification) in 2 subjects, and becoming undetectable in 1 subject.

Discussion

In this small initial study (N = 9), we found no significant differences between pre- and post-chemotherapy levels of HIV-1 RNA in plasma and HIV-1 DNA in PBMC from patients receiving chemotherapy for HIV-1 related lymphoma. The absence of a durable effect on plasma viremia following chemotherapy suggests that there was not a reduction in the number of infected cells that can produce virus. Although much of the HIV-1 DNA that persists despite ART has deletions or is hypermutated [20][22], the lack of a reduction in HIV-1 DNA levels is also consistent with chemotherapy not causing a sustained reduction in infected cell number.

One reason why chemotherapy would not impact HIV-1 persistence is that some subpopulations of CD4+ T cells may be resistant to chemotherapeutic agents. In this regard, Turtle et al. have described a population of CD8+ T cells in peripheral blood that survive intensive chemotherapy [23], and Casorati et al. have described a population of bone marrow resident CD4+ T cells that survive conditioning chemotherapy for autologous transplantation [24]. Importantly, the latent reservoir of HIV-1 resides within the resting memory CD4+ T cell population [3], and while chemotherapy significantly depletes CD4+ T cells in the periphery [8], [24], resting memory CD4+ T cells may be more resistant to cytotoxic chemotherapy because of their quiescent state [15]. In addition, CD4+ T cells that survive cytoreductive chemotherapy and harbor HIV-1 DNA are likely to proliferate in response to chemotherapy-induced lymphopenia through IL-7 mediated homoestatic proliferation [25], [26]. Hence, although chemotherapy may kill some HIV-infected cells, those that survive could repopulate HIV-1 reservoirs through cell proliferation in response to lymphopenia.

Limitations of our study are the small sample size and long interval (median 285 days) between pre- and post-chemotherapy samples. As a consequence, transient reductions in low-level viremia and HIV-infected CD4+ T cells from chemotherapy could have been missed, as could have restoration of viremia and HIV-infected cells through proliferation of surviving CD4+ T cells. Nevertheless, the findings from this current study are similar to those found in a cross-sectional study of 10 patients on suppressive ART, where HIV-1 RNA in plasma and HIV-1 DNA in PBMC remained detectable following myeloablative chemotherapy and autologous stem cell transplantation [13]. The current study adds to this prior post-transplant cross-sectional study by comparing pre- and post-chemotherapy levels of HIV-1 persistence.

The failure of either moderately intensive or myeloablative chemotherapy to have a sustained effect on HIV-1 persistence points to the importance of allogeneic transplantation with a CCR5 delta32 homozygous donor in achieving the first definitive cure of HIV-1 infection [6]. For HIV-1 to infect target cells, CD4 and one of two major coreceptors, either CCR5 or CXCR4, must be expressed on the cell surface [27], [28]. A 32 base pair deletion (CCR5 delta32) provides resistance to CCR5 tropic HIV-1 [29], [30], and is present in 2–5% of persons from Europe, the Middle East and the Indian subcontinent [31]. Complete replacement of the Berlin patient's immune system by allogeneic cells with the CCR5 delta32 mutation was likely critical in eradicating HIV-1 reservoirs. Recently, the elimination of HIV-1 DNA from PBMC in 2 patients who received ART throughout reduced-intensity allogeneic transplantation with CCR5 wild-type donors has been reported [7], confirming the importance of allogeneic transplantation in eliminating evidence of HIV-1 persistence in blood. The recent report of viral rebound in both of these patients 3–8 months after cessation of ART indicates that HIV-1 reservoirs were not eliminated by allogeneic transplantation and that the CCR5 wild-type donor cells supported HIV-1 replication in the absence of ART [32].

In summary, this study provides evidence that chemotherapy alone does not have a sustained impact on HIV-1 persistence in patients on ART and that future therapeutic interventions to reduce or eliminate HIV-1 reservoirs will need to have greater specificity for HIV-infected cells.

Acknowledgments

We are grateful to AIDS Clinical Trial Unit personnel and the patients in ALLRT/A5001 for their participation. We also thank Elizabeth Fyne for her careful review of this manuscript.

Author Contributions

Conceived and designed the experiments: ARC SK DKM RTM MFP JWM. Performed the experiments: ARC. Analyzed the data: ARC SK JWM. Contributed reagents/materials/analysis tools: ARC SK DKM RTM MFP JWM. Wrote the paper: ARC JWM.

References

  1. 1. Maldarelli F, Palmer S, King MS, Wiegand A, Polis MA, et al. (2007) ART suppresses plasma HIV-1 RNA to a stable set point predicted by pretherapy viremia. PLoS Pathog 3: e46.
  2. 2. Palmer S, Maldarelli F, Wiegand A, Bernstein B, Hanna GJ, et al. (2008) Low-level viremia persists for at least 7 years in patients on suppressive antiretroviral therapy. Proc Natl Acad Sci U S A 105: 3879–3884.
  3. 3. Chun TW, Carruth L, Finzi D, Shen X, DiGiuseppe JA, et al. (1997) Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature 387: 183–188.
  4. 4. Wong JK, Hezareh M, Günthard HF, Havlir DV, Ignacio CC, et al. (1997) Recovery of replication-competent HIV despite prolonged suppression of plasma viremia. Science 278: 1291–1295.
  5. 5. Siliciano JD, Kajdas J, Finzi D, Quinn TC, Chadwick K, et al. (2003) Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nat Med 9: 727–728.
  6. 6. Hütter G, Nowak D, Mossner M, Ganepola S, Müssig A, et al. (2009) Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation. N Engl J Med 360: 692–698.
  7. 7. Henrich TJ, Hu Z, Li JZ, Sciaranghella G, Busch MP, et al. (2013) Long-term reduction in peripheral blood HIV type 1 reservoirs following reduced-intensity conditioning allogeneic stem cell transplantation. J Infect Dis 207: 1694–1702.
  8. 8. Petz LD, Redei I, Bryson Y, Regan D, Kurtzberg J, et al. (2013) Hematopoietic cell transplantation with cord blood for cure of HIV infections. Biol Blood Marrow Transplant 19: 393–397.
  9. 9. Tebas P, Stein D, Binder-Scholl G, Mukherjee R, Brady T, et al. (2013) Antiviral effects of autologous CD4 T cells genetically modified with a conditionally replicating lentiviral vector expressing long anti-sense to HIV. Blood 121: 1524–1533.
  10. 10. DiGiusto DL, Krishnan A, Li L, Li H, Li S, et al. (2010) RNA-based gene therapy for HIV with lentiviral vector-modified CD34(+) cells in patients undergoing transplantation for AIDS-related lymphoma. Sci Transl Med 2: 36ra43.
  11. 11. Mitsuyasu RT, Merigan TC, Carr A, Zack JA, Winters MA, et al. (2009) Phase 2 gene therapy trial of an anti-HIV ribozyme in autologous CD34+ cells. Nat Med 15: 285–292.
  12. 12. Zou S, Glynn S, Kuritzkes D, Shah M, Cook N, et al. (2013) Hematopoietic cell transplantation and HIV cure: where we are and what next? Blood 122: 3111–3115.
  13. 13. Cillo AR, Krishnan A, Mitsuyasu RT, McMahon DK, Li S, et al. (2013) Plasma viremia and cellular HIV-1 DNA persist despite autologous hematopoietic stem cell transplantation for HIV-related lymphoma. J Acquir Immune Defic Syndr 63: 438–41.
  14. 14. Mackall CL, Fleisher TA, Brown MR, Andrich MP, Chen CC, et al. (1995) Age, thymopoiesis, and CD4+ T-lymphocyte regeneration after intensive chemotherapy. N Engl J Med 332: 143–149.
  15. 15. Mackall CL, Fleisher TA, Brown MR, Magrath IT, Shad AT, et al. (1994) Lymphocyte depletion during treatment with intensive chemotherapy for cancer. Blood 84: 2221–2228.
  16. 16. Lehrnbecher T, Foster C, Vázquez N, Mackall CL, Chanock SJ (1997) Therapy-induced alterations in host defense in children receiving therapy for cancer. J Pediatr Hematol Oncol 19: 399–417.
  17. 17. Smurzynski M, Collier AC, Koletar SL, Bosch RJ, Wu K, et al. (2008) AIDS clinical trials group longitudinal linked randomized trials (ALLRT): rationale, design, and baseline characteristics. HIV Clin Trials 9: 269–282.
  18. 18. Palmer S, Wiegand AP, Maldarelli F, Bazmi H, Mican JM, et al. (2003) New real-time reverse transcriptase-initiated PCR assay with single-copy sensitivity for human immunodeficiency virus type 1 RNA in plasma. J Clin Microbiol 41: 4531–4536.
  19. 19. Malnati MS, Scarlatti G, Gatto F, Salvatori F, Cassina G, et al. (2008) A universal real-time PCR assay for the quantification of group-M HIV-1 proviral load. Nat Protoc 3: 1240–1248.
  20. 20. Sanchez G, Xu X, Chermann JC, Hirsch I (1997) Accumulation of defective viral genomes in peripheral blood mononuclear cells of human immunodeficiency virus type 1-infected individuals. J Virol 71: 2233–2240.
  21. 21. Mangeat B, Turelli P, Caron G, Friedli M, Perrin L, et al. (2003) Broad antiretroviral defense by human APOBEC3G through lethal editing of nascent reverse transcripts. Nature 424: 99–103.
  22. 22. Ho YC, Shan L, Hosmane NN, Wang J, Laskey SB, et al. (2013) Replication-competent noninduced proviruses in the latent reservoir increase barrier to HIV-1 cure. Cell 155: 540–551.
  23. 23. Turtle CJ, Swanson HM, Fujii N, Estey EH, Riddell SR (2009) A distinct subset of self-renewing human memory CD8+ T cells survives cytotoxic chemotherapy. Immunity 31: 834–844.
  24. 24. Casorati G, Locatelli F, Pagani S, Garavaglia C, Montini E, et al. (2005) Bone marrow-resident memory T cells survive pretransplant chemotherapy and contribute to early immune reconstitution of patients with acute myeloid leukemia given mafosfamide-purged autologous bone marrow transplantation. Exp Hematol 33: 212–218.
  25. 25. Bolotin E, Annett G, Parkman R, Weinberg K (1999) Serum levels of IL-7 in bone marrow transplant recipients: relationship to clinical characteristics and lymphocyte count. Bone Marrow Transplant 23: 783–788.
  26. 26. Chomont N, El-Far M, Ancuta P, Trautmann L, Procopio FA, et al. (2009) HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat Med 15: 893–900.
  27. 27. Feng Y, Broder CC, Kennedy PE, Berger EA (1996) HIV-1 Entry Cofactor: Functional cDNA Cloning of a Seven-Transmembrane, G Protein-Coupled Receptor. Science 272: 872–877.
  28. 28. Alkhatib G, Combadiere C, Broder CC, Feng Y, Kennedy PE, et al. (1996) CC CKR5: A RANTES, MIP-1α, MIP-1β Receptor as a Fusion Cofactor for Macrophage-Tropic HIV-1. Science 272: 1955–1958.
  29. 29. Liu R, Paxton WA, Choe S, Ceradini D, Martin SR, et al. (1996) Homozygous Defect in HIV-1 Coreceptor Accounts for Resistance of Some Multiply-Exposed Individuals to HIV-1 Infection. Cell 86: 367–377.
  30. 30. Agrawal L, Lu X, Qingwen J, VanHorn-Ali Z, Nicolescu IV, et al. (2004) Role for CCR5Δ32 Protein in Resistance to R5, R5X4, and X4 Human Immunodeficiency Virus Type 1 in Primary CD4+ Cells. J Virol 78: 2277–2287.
  31. 31. Martinson JJ, Chapman NH, Rees DC, Liu YT, Clegg JB (1997) Global distribution of CCR5 gene 32-basepair deletion. Nat Genet 16: 100–103.
  32. 32. Henrich, TJ. Challenges and Strategies Towards Functional Cure: How Low Do You Need To Go [abstract]. In: Program and Abstract Book from the Sixth International Workshop on HIV Persistence During Therapy;2013 Dec 3–6; Miami, FL.