Role of Dendritic Cells in Exposing Latent HIV-1 for the Kill
Abstract
:1. Introduction
2. Dendritic Cell Programming for Immunotherapy
2.1. Classification of Endogenous DCs
2.2. Characterization of Ex Vivo-Programmed DCs
3. Use of DCs to Address Hurdles to the CTL ‘Kill’ of HIV-1
3.1. Mutations in CTL Epitopes
3.2. Dysfunctional or Exhausted CTLs
3.3. Defective HIV as Antigen Decoys
3.4. Spatial Separation of CTL and Target Cells
4. Driving HIV-1 out of Hiding: Current Status of Latency Reversal Approaches
4.1. Common LRA Strategies
4.2. Next Generation Pharmacological LRAs?
5. Dual Role for DCs in the ‘Kick and Kill’?
5.1. DCs as a Therapeutic Tool to Drive HIV-1-Specific Killer T cells
5.2. LRA Potential of DCs
5.3. Using DCs to Expose Pathogen-Specific HIV Reservoirs?
5.4. DC Potential as an All-in-One ‘Kick and Kill’ Tool
6. Concluding Remarks
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Barre-Sinoussi, F.; Chermann, J.C.; Rey, F.; Nugeyre, M.T.; Chamaret, S.; Gruest, J.; Dauguet, C.; Axler-Blin, C.; Vezinet-Brun, F.; Rouzioux, C.; et al. Isolation of a T-lymphotropic retrovirus from a patient at risk for acquired immune deficiency syndrome (AIDS). Science 1983, 220, 868–871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- UNAIDS. Global AIDS Update 2018. Available online: https://www.unaids.org/en/20180718_GR2018 (accessed on 19 November 2019).
- UNAIDS. UNAIDS Data 2018. Available online: https://www.unaids.org/sites/default/files/media_asset/unaids-data-2018_en.pdf (accessed on 19 November 2019).
- Ruelas, D.S.; Greene, W.C. An integrated overview of HIV-1 latency. Cell 2013, 155, 519–529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Finzi, D.; Blankson, J.; Siliciano, J.D.; Margolick, J.B.; Chadwick, K.; Pierson, T.; Smith, K.; Lisziewicz, J.; Lori, F.; Flexner, C.; et al. Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy. Nat. Med. 1999, 5, 512. [Google Scholar] [CrossRef] [PubMed]
- Chun, T.W.; Engel, D.; Berrey, M.M.; Shea, T.; Corey, L.; Fauci, A.S. Early establishment of a pool of latently infected, resting CD4+ T cells during primary HIV-1 infection. Proc. Natl. Acad. Sci. USA 1998, 95, 8869–8873. [Google Scholar] [CrossRef] [Green Version]
- Perelson, A.S.; Essunger, P.; Cao, Y.; Vesanen, M.; Hurley, A.; Saksela, K.; Markowitz, M.; Ho, D.D. Decay characteristics of HIV-1-infected compartments during combination therapy. Nature 1997, 387, 188–191. [Google Scholar] [CrossRef]
- Davey, R.T., Jr.; Bhat, N.; Yoder, C.; Chun, T.W.; Metcalf, J.A.; Dewar, R.; Natarajan, V.; Lempicki, R.A.; Adelsberger, J.W.; Miller, K.D.; et al. HIV-1 and T cell dynamics after interruption of highly active antiretroviral therapy (HAART) in patients with a history of sustained viral suppression. Proc. Natl. Acad. Sci. USA 1999, 96, 15109–15114. [Google Scholar] [CrossRef] [Green Version]
- Siliciano, J.D.; Kajdas, J.; Finzi, D.; Quinn, T.C.; Chadwick, K.; Margolick, J.B.; Kovacs, C.; Gange, S.J.; Siliciano, R.F. Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nat. Med. 2003, 9, 727. [Google Scholar] [CrossRef]
- Collin, M.; Bigley, V. Human dendritic cell subsets: An update. Immunology 2018, 154, 3–20. [Google Scholar] [CrossRef]
- Steinman, R.M.; Cohn, Z.A. Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution. J. Exp. Med. 1973, 137, 1142–1162. [Google Scholar] [CrossRef]
- Steinman, R.M.; Cohn, Z.A. Identification of a novel cell type in peripheral lymphoid organs of mice. II. Functional properties in vitro. J. Exp. Med. 1974, 139, 380–397. [Google Scholar] [CrossRef] [Green Version]
- Paul, F.; Arkin, Y.; Giladi, A.; Jaitin, D.A.; Kenigsberg, E.; Keren-Shaul, H.; Winter, D.; Lara-Astiaso, D.; Gury, M.; Weiner, A.; et al. Transcriptional Heterogeneity and Lineage Commitment in Myeloid Progenitors. Cell 2015, 163, 1663–1677. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Notta, F.; Zandi, S.; Takayama, N.; Dobson, S.; Gan, O.I.; Wilson, G.; Kaufmann, K.B.; McLeod, J.; Laurenti, E.; Dunant, C.F.; et al. Distinct routes of lineage development reshape the human blood hierarchy across ontogeny. Science 2016, 351, aab2116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Velten, L.; Haas, S.F.; Raffel, S.; Blaszkiewicz, S.; Islam, S.; Hennig, B.P.; Hirche, C.; Lutz, C.; Buss, E.C.; Nowak, D.; et al. Human haematopoietic stem cell lineage commitment is a continuous process. Nat. Cell Biol. 2017, 19, 271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karamitros, D.; Stoilova, B.; Aboukhalil, Z.; Hamey, F.; Reinisch, A.; Samitsch, M.; Quek, L.; Otto, G.; Repapi, E.; Doondeea, J.; et al. Single-cell analysis reveals the continuum of human lympho-myeloid progenitor cells. Nat. Immunol. 2018, 19, 85–97. [Google Scholar] [CrossRef] [PubMed]
- Guilliams, M.; Dutertre, C.A.; Scott, C.L.; McGovern, N.; Sichien, D.; Chakarov, S.; Van Gassen, S.; Chen, J.; Poidinger, M.; De Prijck, S.; et al. Unsupervised High-Dimensional Analysis Aligns Dendritic Cells across Tissues and Species. Immunity 2016, 45, 669–684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guilliams, M.; Ginhoux, F.; Jakubzick, C.; Naik, S.H.; Onai, N.; Schraml, B.U.; Segura, E.; Tussiwand, R.; Yona, S. Dendritic cells, monocytes and macrophages: A unified nomenclature based on ontogeny. Nat. Rev. Immunol. 2014, 14, 571–578. [Google Scholar] [CrossRef] [PubMed]
- Heidkamp, G.F.; Sander, J.; Lehmann, C.H.K.; Heger, L.; Eissing, N.; Baranska, A.; Luhr, J.J.; Hoffmann, A.; Reimer, K.C.; Lux, A.; et al. Human lymphoid organ dendritic cell identity is predominantly dictated by ontogeny, not tissue microenvironment. Sci. Immunol. 2016, 1, eaai7677. [Google Scholar] [CrossRef]
- Granot, T.; Senda, T.; Carpenter, D.J.; Matsuoka, N.; Weiner, J.; Gordon, C.L.; Miron, M.; Kumar, B.V.; Griesemer, A.; Ho, S.H.; et al. Dendritic Cells Display Subset and Tissue-Specific Maturation Dynamics over Human Life. Immunity 2017, 46, 504–515. [Google Scholar] [CrossRef] [Green Version]
- Bao, M.; Liu, Y.J. Regulation of TLR7/9 signaling in plasmacytoid dendritic cells. Protein Cell 2013, 4, 40–52. [Google Scholar] [CrossRef] [Green Version]
- Swiecki, M.; Colonna, M. The multifaceted biology of plasmacytoid dendritic cells. Nat. Rev. Immunol. 2015, 15, 471–485. [Google Scholar] [CrossRef]
- Dzionek, A.; Fuchs, A.; Schmidt, P.; Cremer, S.; Zysk, M.; Miltenyi, S.; Buck, D.W.; Schmitz, J. BDCA-2, BDCA-3, and BDCA-4: Three markers for distinct subsets of dendritic cells in human peripheral blood. J. Immunol. 2000, 165, 6037–6046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- MacDonald, K.P.; Munster, D.J.; Clark, G.J.; Dzionek, A.; Schmitz, J.; Hart, D.N. Characterization of human blood dendritic cell subsets. Blood 2002, 100, 4512–4520. [Google Scholar] [CrossRef] [PubMed]
- Ahrens, S.; Zelenay, S.; Sancho, D.; Hanc, P.; Kjaer, S.; Feest, C.; Fletcher, G.; Durkin, C.; Postigo, A.; Skehel, M.; et al. F-actin is an evolutionarily conserved damage-associated molecular pattern recognized by DNGR-1, a receptor for dead cells. Immunity 2012, 36, 635–645. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, J.G.; Czabotar, P.E.; Policheni, A.N.; Caminschi, I.; Wan, S.S.; Kitsoulis, S.; Tullett, K.M.; Robin, A.Y.; Brammananth, R.; van Delft, M.F.; et al. The dendritic cell receptor Clec9A binds damaged cells via exposed actin filaments. Immunity 2012, 36, 646–657. [Google Scholar] [CrossRef] [Green Version]
- Haniffa, M.; Shin, A.; Bigley, V.; McGovern, N.; Teo, P.; See, P.; Wasan, P.S.; Wang, X.N.; Malinarich, F.; Malleret, B.; et al. Human tissues contain CD141hi cross-presenting dendritic cells with functional homology to mouse CD103+ nonlymphoid dendritic cells. Immunity 2012, 37, 60–73. [Google Scholar] [CrossRef] [Green Version]
- Poulin, L.F.; Salio, M.; Griessinger, E.; Anjos-Afonso, F.; Craciun, L.; Chen, J.L.; Keller, A.M.; Joffre, O.; Zelenay, S.; Nye, E.; et al. Characterization of human DNGR-1+ BDCA3+ leukocytes as putative equivalents of mouse CD8alpha+ dendritic cells. J. Exp. Med. 2010, 207, 1261–1271. [Google Scholar] [CrossRef]
- Jongbloed, S.L.; Kassianos, A.J.; McDonald, K.J.; Clark, G.J.; Ju, X.; Angel, C.E.; Chen, C.J.; Dunbar, P.R.; Wadley, R.B.; Jeet, V.; et al. Human CD141+ (BDCA-3)+ dendritic cells (DCs) represent a unique myeloid DC subset that cross-presents necrotic cell antigens. J. Exp. Med. 2010, 207, 1247–1260. [Google Scholar] [CrossRef] [Green Version]
- Bachem, A.; Guttler, S.; Hartung, E.; Ebstein, F.; Schaefer, M.; Tannert, A.; Salama, A.; Movassaghi, K.; Opitz, C.; Mages, H.W.; et al. Superior antigen cross-presentation and XCR1 expression define human CD11c+CD141+ cells as homologues of mouse CD8+ dendritic cells. J. Exp. Med. 2010, 207, 1273–1281. [Google Scholar] [CrossRef] [Green Version]
- Hemont, C.; Neel, A.; Heslan, M.; Braudeau, C.; Josien, R. Human blood mDC subsets exhibit distinct TLR repertoire and responsiveness. J. Leukoc. Biol. 2013, 93, 599–609. [Google Scholar] [CrossRef]
- Colletti, N.J.; Liu, H.; Gower, A.C.; Alekseyev, Y.O.; Arendt, C.W.; Shaw, M.H. TLR3 Signaling Promotes the Induction of Unique Human BDCA-3 Dendritic Cell Populations. Front. Immunol. 2016, 7, 88. [Google Scholar] [CrossRef] [Green Version]
- Lauterbach, H.; Bathke, B.; Gilles, S.; Traidl-Hoffmann, C.; Luber, C.A.; Fejer, G.; Freudenberg, M.A.; Davey, G.M.; Vremec, D.; Kallies, A.; et al. Mouse CD8alpha+ DCs and human BDCA3+ DCs are major producers of IFN-lambda in response to poly IC. J. Exp. Med. 2010, 207, 2703–2717. [Google Scholar] [CrossRef]
- Sittig, S.P.; Bakdash, G.; Weiden, J.; Skold, A.E.; Tel, J.; Figdor, C.G.; de Vries, I.J.; Schreibelt, G. A Comparative Study of the T Cell Stimulatory and Polarizing Capacity of Human Primary Blood Dendritic Cell Subsets. Mediat. Inflamm. 2016, 2016, 3605643. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nizzoli, G.; Larghi, P.; Paroni, M.; Crosti, M.C.; Moro, M.; Neddermann, P.; Caprioli, F.; Pagani, M.; De Francesco, R.; Abrignani, S.; et al. IL-10 promotes homeostatic proliferation of human CD8+ memory T cells and, when produced by CD1c+ DCs, shapes naive CD8+ T-cell priming. Eur. J. Immunol. 2016, 46, 1622–1632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Di Blasio, S.; Wortel, I.M.; van Bladel, D.A.; de Vries, L.E.; Duiveman-de Boer, T.; Worah, K.; de Haas, N.; Buschow, S.I.; de Vries, I.J.; Figdor, C.G.; et al. Human CD1c+ DCs are critical cellular mediators of immune responses induced by immunogenic cell death. Oncoimmunology 2016, 5, e1192739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mittag, D.; Proietto, A.I.; Loudovaris, T.; Mannering, S.I.; Vremec, D.; Shortman, K.; Wu, L.; Harrison, L.C. Human dendritic cell subsets from spleen and blood are similar in phenotype and function but modified by donor health status. J. Immunol. 2011, 186, 6207–6217. [Google Scholar] [CrossRef] [Green Version]
- Segura, E.; Durand, M.; Amigorena, S. Similar antigen cross-presentation capacity and phagocytic functions in all freshly isolated human lymphoid organ-resident dendritic cells. J. Exp. Med. 2013, 210, 1035–1047. [Google Scholar] [CrossRef] [Green Version]
- Nizzoli, G.; Krietsch, J.; Weick, A.; Steinfelder, S.; Facciotti, F.; Gruarin, P.; Bianco, A.; Steckel, B.; Moro, M.; Crosti, M.; et al. Human CD1c+ dendritic cells secrete high levels of IL-12 and potently prime cytotoxic T-cell responses. Blood 2013, 122, 932–942. [Google Scholar] [CrossRef] [Green Version]
- Cohn, L.; Chatterjee, B.; Esselborn, F.; Smed-Sorensen, A.; Nakamura, N.; Chalouni, C.; Lee, B.C.; Vandlen, R.; Keler, T.; Lauer, P.; et al. Antigen delivery to early endosomes eliminates the superiority of human blood BDCA3+ dendritic cells at cross presentation. J. Exp. Med. 2013, 210, 1049–1063. [Google Scholar] [CrossRef] [Green Version]
- Bell, D.; Chomarat, P.; Broyles, D.; Netto, G.; Harb, G.M.; Lebecque, S.; Valladeau, J.; Davoust, J.; Palucka, K.A.; Banchereau, J. In breast carcinoma tissue, immature dendritic cells reside within the tumor, whereas mature dendritic cells are located in peritumoral areas. J. Exp. Med. 1999, 190, 1417–1426. [Google Scholar] [CrossRef] [Green Version]
- Toriyama, K.; Wen, D.R.; Paul, E.; Cochran, A.J. Variations in the distribution, frequency, and phenotype of Langerhans cells during the evolution of malignant melanoma of the skin. J. Investig. Dermatol. 1993, 100, S269–S273. [Google Scholar] [CrossRef] [Green Version]
- Troy, A.J.; Summers, K.L.; Davidson, P.J.; Atkinson, C.H.; Hart, D.N. Minimal recruitment and activation of dendritic cells within renal cell carcinoma. Clin. Cancer Res. 1998, 4, 585–593. [Google Scholar] [PubMed]
- Zou, W.; Machelon, V.; Coulomb-L’Hermin, A.; Borvak, J.; Nome, F.; Isaeva, T.; Wei, S.; Krzysiek, R.; Durand-Gasselin, I.; Gordon, A.; et al. Stromal-derived factor-1 in human tumors recruits and alters the function of plasmacytoid precursor dendritic cells. Nat. Med. 2001, 7, 1339–1346. [Google Scholar] [CrossRef] [PubMed]
- Almand, B.; Resser, J.R.; Lindman, B.; Nadaf, S.; Clark, J.I.; Kwon, E.D.; Carbone, D.P.; Gabrilovich, D.I. Clinical significance of defective dendritic cell differentiation in cancer. Clin. Cancer Res. 2000, 6, 1755–1766. [Google Scholar] [PubMed]
- Della Bella, S.; Gennaro, M.; Vaccari, M.; Ferraris, C.; Nicola, S.; Riva, A.; Clerici, M.; Greco, M.; Villa, M.L. Altered maturation of peripheral blood dendritic cells in patients with breast cancer. Br. J. Cancer 2003, 89, 1463–1472. [Google Scholar] [CrossRef] [Green Version]
- Kalinski, P.; Edington, H.; Zeh, H.J.; Okada, H.; Butterfield, L.H.; Kirkwood, J.M.; Bartlett, D.L. Dendritic cells in cancer immunotherapy: Vaccines or autologous transplants? Immunol. Res. 2011, 50, 235–247. [Google Scholar] [CrossRef] [Green Version]
- Amigorena, S.; Savina, A. Intracellular mechanisms of antigen cross presentation in dendritic cells. Curr. Opin. Immunol. 2010, 22, 109–117. [Google Scholar] [CrossRef]
- Segura, E.; Villadangos, J.A. Antigen presentation by dendritic cells in vivo. Curr. Opin. Immunol. 2009, 21, 105–110. [Google Scholar] [CrossRef]
- Shortman, K.; Heath, W.R. The CD8+ dendritic cell subset. Immunol. Rev. 2010, 234, 18–31. [Google Scholar] [CrossRef]
- Manfredi, A.A.; Capobianco, A.; Bianchi, M.E.; Rovere-Querini, P. Regulation of dendritic- and T-cell fate by injury-associated endogenous signals. Crit. Rev. Immunol. 2009, 29, 69–86. [Google Scholar] [CrossRef]
- Lopez-Albaitero, A.; Mailliard, R.; Hackman, T.; Andrade Filho, P.A.; Wang, X.; Gooding, W.; Ferrone, S.; Kalinski, P.; Ferris, R.L. Maturation pathways of dendritic cells determine TAP1 and TAP2 levels and cross-presenting function. J. Immunother. 2009, 32, 465. [Google Scholar] [CrossRef] [Green Version]
- De Vries, I.J.; Krooshoop, D.J.; Scharenborg, N.M.; Lesterhuis, W.J.; Diepstra, J.H.; Van Muijen, G.N.; Strijk, S.P.; Ruers, T.J.; Boerman, O.C.; Oyen, W.J.; et al. Effective migration of antigen-pulsed dendritic cells to lymph nodes in melanoma patients is determined by their maturation state. Cancer Res. 2003, 63, 12–17. [Google Scholar] [PubMed]
- De Vries, I.J.; Lesterhuis, W.J.; Scharenborg, N.M.; Engelen, L.P.; Ruiter, D.J.; Gerritsen, M.J.; Croockewit, S.; Britten, C.M.; Torensma, R.; Adema, G.J.; et al. Maturation of dendritic cells is a prerequisite for inducing immune responses in advanced melanoma patients. Clin. Cancer Res. 2003, 9, 5091–5100. [Google Scholar] [PubMed]
- Dhodapkar, M.V.; Steinman, R.M.; Sapp, M.; Desai, H.; Fossella, C.; Krasovsky, J.; Donahoe, S.M.; Dunbar, P.R.; Cerundolo, V.; Nixon, D.F.; et al. Rapid generation of broad T-cell immunity in humans after a single injection of mature dendritic cells. J. Clin. Investig. 1999, 104, 173–180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Adema, G.J.; de Vries, I.J.; Punt, C.J.; Figdor, C.G. Migration of dendritic cell based cancer vaccines: In vivo veritas? Curr. Opin. Immunol. 2005, 17, 170–174. [Google Scholar] [CrossRef] [PubMed]
- Bender, A.; Sapp, M.; Schuler, G.; Steinman, R.M.; Bhardwaj, N. Improved methods for the generation of dendritic cells from nonproliferating progenitors in human blood. J. Immunol. Methods 1996, 196, 121–135. [Google Scholar] [CrossRef]
- Reddy, A.; Sapp, M.; Feldman, M.; Subklewe, M.; Bhardwaj, N. A monocyte conditioned medium is more effective than defined cytokines in mediating the terminal maturation of human dendritic cells. Blood 1997, 90, 3640–3646. [Google Scholar] [CrossRef]
- Jonuleit, H.; Kuhn, U.; Muller, G.; Steinbrink, K.; Paragnik, L.; Schmitt, E.; Knop, J.; Enk, A.H. Pro-inflammatory cytokines and prostaglandins induce maturation of potent immunostimulatory dendritic cells under fetal calf serum-free conditions. Eur. J. Immunol. 1997, 27, 3135–3142. [Google Scholar] [CrossRef]
- Luft, T.; Jefford, M.; Luetjens, P.; Toy, T.; Hochrein, H.; Masterman, K.A.; Maliszewski, C.; Shortman, K.; Cebon, J.; Maraskovsky, E. Functionally distinct dendritic cell (DC) populations induced by physiologic stimuli: Prostaglandin E2 regulates the migratory capacity of specific DC subsets. Blood 2002, 100, 1362–1372. [Google Scholar] [CrossRef] [Green Version]
- Scandella, E.; Men, Y.; Gillessen, S.; Forster, R.; Groettrup, M. Prostaglandin E2 is a key factor for CCR7 surface expression and migration of monocyte-derived dendritic cells. Blood 2002, 100, 1354–1361. [Google Scholar] [CrossRef]
- Mailliard, R.B.; Wankowicz-Kalinska, A.; Cai, Q.; Wesa, A.; Hilkens, C.M.; Kapsenberg, M.L.; Kirkwood, J.M.; Storkus, W.J.; Kalinski, P. alpha-type-1 polarized dendritic cells: A novel immunization tool with optimized CTL-inducing activity. Cancer Res. 2004, 64, 5934–5937. [Google Scholar] [CrossRef] [Green Version]
- Muthuswamy, R.; Mueller-Berghaus, J.; Haberkorn, U.; Reinhart, T.A.; Schadendorf, D.; Kalinski, P. PGE2 transiently enhances DC expression of CCR7 but inhibits the ability of DCs to produce CCL19 and attract naive T cells. Blood 2010, 116, 1454–1459. [Google Scholar] [CrossRef] [PubMed]
- Zaccard, C.R.; Watkins, S.C.; Kalinski, P.; Fecek, R.J.; Yates, A.L.; Salter, R.D.; Ayyavoo, V.; Rinaldo, C.R.; Mailliard, R.B. CD40L induces functional tunneling nanotube networks exclusively in dendritic cells programmed by mediators of type 1 immunity. J. Immunol. 2015, 194, 1047–1056. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kubo, S.; Takahashi, H.K.; Takei, M.; Iwagaki, H.; Yoshino, T.; Tanaka, N.; Mori, S.; Nishibori, M. E-prostanoid (EP)2/EP4 receptor-dependent maturation of human monocyte-derived dendritic cells and induction of helper T2 polarization. J. Pharmacol. Exp. Ther. 2004, 309, 1213–1220. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kalinski, P.; Schuitemaker, J.H.; Hilkens, C.M.; Kapsenberg, M.L. Prostaglandin E2 induces the final maturation of IL-12-deficient CD1a+CD83+ dendritic cells: The levels of IL-12 are determined during the final dendritic cell maturation and are resistant to further modulation. J. Immunol. 1998, 161, 2804–2809. [Google Scholar] [PubMed]
- Vieira, P.L.; de Jong, E.C.; Wierenga, E.A.; Kapsenberg, M.L.; Kalinski, P. Development of Th1-inducing capacity in myeloid dendritic cells requires environmental instruction. J. Immunol. 2000, 164, 4507–4512. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kalinski, P.; Vieira, P.L.; Schuitemaker, J.H.; de Jong, E.C.; Kapsenberg, M.L. Prostaglandin E2 is a selective inducer of interleukin-12 p40 (IL-12p40) production and an inhibitor of bioactive IL-12p70 heterodimer. Blood 2001, 97, 3466–3469. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Muthuswamy, R.; Urban, J.; Lee, J.J.; Reinhart, T.A.; Bartlett, D.; Kalinski, P. Ability of mature dendritic cells to interact with regulatory T cells is imprinted during maturation. Cancer Res. 2008, 68, 5972–5978. [Google Scholar] [CrossRef] [Green Version]
- Schadendorf, D.; Ugurel, S.; Schuler-Thurner, B.; Nestle, F.O.; Enk, A.; Brocker, E.B.; Grabbe, S.; Rittgen, W.; Edler, L.; Sucker, A.; et al. Dacarbazine (DTIC) versus vaccination with autologous peptide-pulsed dendritic cells (DC) in first-line treatment of patients with metastatic melanoma: A randomized phase III trial of the DC study group of the DeCOG. Ann. Oncol. 2006, 17, 563–570. [Google Scholar] [CrossRef]
- Macatangay, B.J.; Szajnik, M.E.; Whiteside, T.L.; Riddler, S.A.; Rinaldo, C.R. Regulatory T cell suppression of Gag-specific CD8 T cell polyfunctional response after therapeutic vaccination of HIV-1-infected patients on ART. PLoS ONE 2010, 5, e9852. [Google Scholar] [CrossRef] [Green Version]
- Wesa, A.; Kalinski, P.; Kirkwood, J.M.; Tatsumi, T.; Storkus, W.J. Polarized type-1 dendritic cells (DC1) producing high levels of IL-12 family members rescue patient TH1-type antimelanoma CD4+ T cell responses in vitro. J. Immunother. 2007, 30, 75–82. [Google Scholar] [CrossRef]
- Gustafsson, K.; Junevik, K.; Werlenius, O.; Holmgren, S.; Karlsson-Parra, A.; Andersson, P.O. Tumour-loaded alpha-type 1-polarized dendritic cells from patients with chronic lymphocytic leukaemia produce a superior NK-, NKT- and CD8+ T cell-attracting chemokine profile. Scand. J. Immunol. 2011, 74, 318–326. [Google Scholar] [CrossRef] [PubMed]
- Mailliard, R.B.; Smith, K.N.; Fecek, R.J.; Rappocciolo, G.; Nascimento, E.J.; Marques, E.T.; Watkins, S.C.; Mullins, J.I.; Rinaldo, C.R. Selective induction of CTL helper rather than killer activity by natural epitope variants promotes dendritic cell-mediated HIV-1 dissemination. J. Immunol. 2013, 191, 2570–2580. [Google Scholar] [CrossRef] [PubMed]
- Zitvogel, L.; Mayordomo, J.I.; Tjandrawan, T.; DeLeo, A.B.; Clarke, M.R.; Lotze, M.T.; Storkus, W.J. Therapy of murine tumors with tumor peptide-pulsed dendritic cells: Dependence on T cells, B7 costimulation, and T helper cell 1-associated cytokines. J. Exp. Med. 1996, 183, 87–97. [Google Scholar] [CrossRef] [PubMed]
- Zitvogel, L.; Robbins, P.D.; Storkus, W.J.; Clarke, M.R.; Maeurer, M.J.; Campbell, R.L.; Davis, C.G.; Tahara, H.; Schreiber, R.D.; Lotze, M.T. Interleukin-12 and B7.1 co-stimulation cooperate in the induction of effective antitumor immunity and therapy of established tumors. Eur. J. Immunol. 1996, 26, 1335–1341. [Google Scholar] [CrossRef]
- Furumoto, K.; Arii, S.; Yamasaki, S.; Mizumoto, M.; Mori, A.; Inoue, N.; Isobe, N.; Imamura, M. Spleen-derived dendritic cells engineered to enhance interleukin-12 production elicit therapeutic antitumor immune responses. Int. J. Cancer 2000, 87, 665–672. [Google Scholar] [CrossRef]
- Furumoto, K.; Mori, A.; Yamasaki, S.; Inoue, N.; Yang, W.; Nakau, M.; Yasuda, S.; Arii, S.; Imamura, M. Interleukin-12-gene transduction makes DCs from tumor-bearing mice an effective inducer of tumor-specific immunity in a peritoneal dissemination model. Immunol. Lett. 2002, 83, 13–20. [Google Scholar] [CrossRef]
- Nishioka, Y.; Hirao, M.; Robbins, P.D.; Lotze, M.T.; Tahara, H. Induction of systemic and therapeutic antitumor immunity using intratumoral injection of dendritic cells genetically modified to express interleukin 12. Cancer Res. 1999, 59, 4035–4041. [Google Scholar]
- Okada, H.; Kalinski, P.; Ueda, R.; Hoji, A.; Kohanbash, G.; Donegan, T.E.; Mintz, A.H.; Engh, J.A.; Bartlett, D.L.; Brown, C.K.; et al. Induction of CD8+ T-cell responses against novel glioma-associated antigen peptides and clinical activity by vaccinations with {alpha}-type 1 polarized dendritic cells and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in patients with recurrent malignant glioma. J. Clin. Oncol. 2011, 29, 330–336. [Google Scholar] [CrossRef] [Green Version]
- Redlinger, R.E., Jr.; Mailliard, R.B.; Barksdale, E.M., Jr. Advanced neuroblastoma impairs dendritic cell function in adoptive immunotherapy. J. Pediatr. Surg. 2003, 38, 857–862. [Google Scholar] [CrossRef]
- Satoh, Y.; Esche, C.; Gambotto, A.; Shurin, G.V.; Yurkovetsky, Z.R.; Robbins, P.D.; Watkins, S.C.; Todo, S.; Herberman, R.B.; Lotze, M.T.; et al. Local administration of IL-12-transfected dendritic cells induces antitumor immune responses to colon adenocarcinoma in the liver in mice. J. Exp. Ther. Oncol. 2002, 2, 337–349. [Google Scholar] [CrossRef]
- Shimizu, T.; Berhanu, A.; Redlinger, R.E., Jr.; Watkins, S.; Lotze, M.T.; Barksdale, E.M., Jr. Interleukin-12 transduced dendritic cells induce regression of established murine neuroblastoma. J. Pediatr. Surg. 2001, 36, 1285–1292. [Google Scholar] [CrossRef] [PubMed]
- Yamanaka, R.; Zullo, S.A.; Ramsey, J.; Yajima, N.; Tsuchiya, N.; Tanaka, R.; Blaese, M.; Xanthopoulos, K.G. Marked enhancement of antitumor immune responses in mouse brain tumor models by genetically modified dendritic cells producing Semliki Forest virus-mediated interleukin-12. J. Neurosurg. 2002, 97, 611–618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, S.; Zeng, G.; Wilkes, D.S.; Reed, G.E.; McGarry, R.C.; Eble, J.N.; Cheng, L. Dendritic cells transfected with interleukin-12 and pulsed with tumor extract inhibit growth of murine prostatic carcinoma in vivo. Prostate 2003, 55, 292–298. [Google Scholar] [CrossRef] [PubMed]
- Kalinski, P.; Schuitemaker, J.H.; Hilkens, C.M.; Wierenga, E.A.; Kapsenberg, M.L. Final maturation of dendritic cells is associated with impaired responsiveness to IFN-gamma and to bacterial IL-12 inducers: Decreased ability of mature dendritic cells to produce IL-12 during the interaction with Th cells. J. Immunol. 1999, 162, 3231–3236. [Google Scholar] [PubMed]
- Langenkamp, A.; Messi, M.; Lanzavecchia, A.; Sallusto, F. Kinetics of dendritic cell activation: Impact on priming of TH1, TH2 and nonpolarized T cells. Nat. Immunol. 2000, 1, 311–316. [Google Scholar] [CrossRef]
- Kalinski, P.; Giermasz, A.; Nakamura, Y.; Basse, P.; Storkus, W.J.; Kirkwood, J.M.; Mailliard, R.B. Helper role of NK cells during the induction of anticancer responses by dendritic cells. Mol. Immunol. 2005, 42, 535–539. [Google Scholar] [CrossRef]
- Kalinski, P.; Mailliard, R.B.; Giermasz, A.; Zeh, H.J.; Basse, P.; Bartlett, D.L.; Kirkwood, J.M.; Lotze, M.T.; Herberman, R.B. Natural killer-dendritic cell cross-talk in cancer immunotherapy. Expert Opin. Biol. Ther. 2005, 5, 1303–1315. [Google Scholar] [CrossRef]
- Mailliard, R.B.; Egawa, S.; Cai, Q.; Kalinska, A.; Bykovskaya, S.N.; Lotze, M.T.; Kapsenberg, M.L.; Storkus, W.J.; Kalinski, P. Complementary dendritic cell-activating function of CD8+ and CD4+ T cells: Helper role of CD8+ T cells in the development of T helper type 1 responses. J. Exp. Med. 2002, 195, 473–483. [Google Scholar] [CrossRef] [Green Version]
- Mailliard, R.B.; Son, Y.I.; Redlinger, R.; Coates, P.T.; Giermasz, A.; Morel, P.A.; Storkus, W.J.; Kalinski, P. Dendritic cells mediate NK cell help for Th1 and CTL responses: Two-signal requirement for the induction of NK cell helper function. J. Immunol. 2003, 171, 2366–2373. [Google Scholar] [CrossRef]
- Xu, S.; Koski, G.K.; Faries, M.; Bedrosian, I.; Mick, R.; Maeurer, M.; Cheever, M.A.; Cohen, P.A.; Czerniecki, B.J. Rapid high efficiency sensitization of CD8+ T cells to tumor antigens by dendritic cells leads to enhanced functional avidity and direct tumor recognition through an IL-12-dependent mechanism. J. Immunol. 2003, 171, 2251–2261. [Google Scholar] [CrossRef] [Green Version]
- Kalinski, P.; Nakamura, Y.; Watchmaker, P.; Giermasz, A.; Muthuswamy, R.; Mailliard, R.B. Helper roles of NK and CD8+ T cells in the induction of tumor immunity. Polarized dendritic cells as cancer vaccines. Immunol. Res. 2006, 36, 137–146. [Google Scholar] [CrossRef]
- Ten Brinke, A.; Karsten, M.L.; Dieker, M.C.; Zwaginga, J.J.; van Ham, S.M. The clinical grade maturation cocktail monophosphoryl lipid A plus IFNgamma generates monocyte-derived dendritic cells with the capacity to migrate and induce Th1 polarization. Vaccine 2007, 25, 7145–7152. [Google Scholar] [CrossRef] [PubMed]
- Camporeale, A.; Boni, A.; Iezzi, G.; Degl’Innocenti, E.; Grioni, M.; Mondino, A.; Bellone, M. Critical impact of the kinetics of dendritic cells activation on the in vivo induction of tumor-specific T lymphocytes. Cancer Res. 2003, 63, 3688–3694. [Google Scholar] [PubMed]
- Watchmaker, P.B.; Berk, E.; Muthuswamy, R.; Mailliard, R.B.; Urban, J.A.; Kirkwood, J.M.; Kalinski, P. Independent regulation of chemokine responsiveness and cytolytic function versus CD8+ T cell expansion by dendritic cells. J. Immunol. 2010, 184, 591–597. [Google Scholar] [CrossRef] [Green Version]
- Barratt-Boyes, S.M.; Figdor, C.G. Current issues in delivering DCs for immunotherapy. Cytotherapy 2004, 6, 105–110. [Google Scholar] [CrossRef]
- Jusforgues-Saklani, H.; Uhl, M.; Blachere, N.; Lemaitre, F.; Lantz, O.; Bousso, P.; Braun, D.; Moon, J.J.; Albert, M.L. Antigen persistence is required for dendritic cell licensing and CD8+ T cell cross-priming. J. Immunol. 2008, 181, 3067–3076. [Google Scholar] [CrossRef] [Green Version]
- Archin, N.M.; Liberty, A.L.; Kashuba, A.D.; Choudhary, S.K.; Kuruc, J.D.; Crooks, A.M.; Parker, D.C.; Anderson, E.M.; Kearney, M.F.; Strain, M.C.; et al. Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy. Nature 2012, 487, 482–485. [Google Scholar] [CrossRef] [Green Version]
- Archin, N.M.; Kirchherr, J.L.; Sung, J.A.; Clutton, G.; Sholtis, K.; Xu, Y.; Allard, B.; Stuelke, E.; Kashuba, A.D.; Kuruc, J.D.; et al. Interval dosing with the HDAC inhibitor vorinostat effectively reverses HIV latency. J. Clin. Investig. 2017, 127, 3126–3135. [Google Scholar] [CrossRef] [Green Version]
- Elliott, J.H.; McMahon, J.H.; Chang, C.C.; Lee, S.A.; Hartogensis, W.; Bumpus, N.; Savic, R.; Roney, J.; Hoh, R.; Solomon, A.; et al. Short-term administration of disulfiram for reversal of latent HIV infection: A phase 2 dose-escalation study. Lancet HIV 2015, 2, e520–e529. [Google Scholar] [CrossRef] [Green Version]
- Elliott, J.H.; Wightman, F.; Solomon, A.; Ghneim, K.; Ahlers, J.; Cameron, M.J.; Smith, M.Z.; Spelman, T.; McMahon, J.; Velayudham, P.; et al. Activation of HIV transcription with short-course vorinostat in HIV-infected patients on suppressive antiretroviral therapy. PLoS Pathog. 2014, 10, e1004473. [Google Scholar] [CrossRef] [Green Version]
- Gutierrez, C.; Serrano-Villar, S.; Madrid-Elena, N.; Perez-Elias, M.J.; Martin, M.E.; Barbas, C.; Ruiperez, J.; Munoz, E.; Munoz-Fernandez, M.A.; Castor, T.; et al. Bryostatin-1 for latent virus reactivation in HIV-infected patients on antiretroviral therapy. Aids 2016, 30, 1385–1392. [Google Scholar] [CrossRef] [PubMed]
- Rasmussen, T.A.; Tolstrup, M.; Brinkmann, C.R.; Olesen, R.; Erikstrup, C.; Solomon, A.; Winckelmann, A.; Palmer, S.; Dinarello, C.; Buzon, M.; et al. Panobinostat, a histone deacetylase inhibitor, for latent-virus reactivation in HIV-infected patients on suppressive antiretroviral therapy: A phase 1/2, single group, clinical trial. Lancet HIV 2014, 1, e13–e21. [Google Scholar] [CrossRef]
- Sogaard, O.S.; Graversen, M.E.; Leth, S.; Olesen, R.; Brinkmann, C.R.; Nissen, S.K.; Kjaer, A.S.; Schleimann, M.H.; Denton, P.W.; Hey-Cunningham, W.J.; et al. The Depsipeptide Romidepsin Reverses HIV-1 Latency In Vivo. PLoS Pathog. 2015, 11, e1005142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Spivak, A.M.; Planelles, V. HIV-1 Eradication: Early Trials (and Tribulations). Trends Mol. Med. 2016, 22, 10–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rasmussen, T.A.; Tolstrup, M.; Sogaard, O.S. Reversal of Latency as Part of a Cure for HIV-1. Trends Microbiol. 2016, 24, 90–97. [Google Scholar] [CrossRef] [PubMed]
- Jin, X.; Bauer, D.E.; Tuttleton, S.E.; Lewin, S.; Gettie, A.; Blanchard, J.; Irwin, C.E.; Safrit, J.T.; Mittler, J.; Weinberger, L.; et al. Dramatic rise in plasma viremia after CD8+ T cell depletion in simian immunodeficiency virus-infected macaques. J. Exp. Med. 1999, 189, 991–998. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schmitz, J.E.; Kuroda, M.J.; Santra, S.; Sasseville, V.G.; Simon, M.A.; Lifton, M.A.; Racz, P.; Tenner-Racz, K.; Dalesandro, M.; Scallon, B.J.; et al. Control of viremia in simian immunodeficiency virus infection by CD8+ lymphocytes. Science 1999, 283, 857–860. [Google Scholar] [CrossRef] [Green Version]
- Chowdhury, A.; Hayes, T.L.; Bosinger, S.E.; Lawson, B.O.; Vanderford, T.; Schmitz, J.E.; Paiardini, M.; Betts, M.; Chahroudi, A.; Estes, J.D.; et al. Differential Impact of In Vivo CD8+ T Lymphocyte Depletion in Controller versus Progressor Simian Immunodeficiency Virus-Infected Macaques. J. Virol. 2015, 89, 8677–8686. [Google Scholar] [CrossRef] [Green Version]
- Cartwright, E.K.; Spicer, L.; Smith, S.A.; Lee, D.; Fast, R.; Paganini, S.; Lawson, B.O.; Nega, M.; Easley, K.; Schmitz, J.E.; et al. CD8+ Lymphocytes Are Required for Maintaining Viral Suppression in SIV-Infected Macaques Treated with Short-Term Antiretroviral Therapy. Immunity 2016, 45, 656–668. [Google Scholar] [CrossRef] [Green Version]
- Cao, Y.; Cartwright, E.K.; Silvestri, G.; Perelson, A.S. CD8+ lymphocyte control of SIV infection during antiretroviral therapy. PLoS Pathog. 2018, 14, e1007350. [Google Scholar] [CrossRef] [Green Version]
- Borrow, P.; Lewicki, H.; Hahn, B.H.; Shaw, G.M.; Oldstone, M.B. Virus-specific CD8+ cytotoxic T-lymphocyte activity associated with control of viremia in primary human immunodeficiency virus type 1 infection. J. Virol. 1994, 68, 6103–6110. [Google Scholar] [PubMed]
- Koup, R.A.; Safrit, J.T.; Cao, Y.; Andrews, C.A.; McLeod, G.; Borkowsky, W.; Farthing, C.; Ho, D.D. Temporal association of cellular immune responses with the initial control of viremia in primary human immunodeficiency virus type 1 syndrome. J. Virol. 1994, 68, 4650–4655. [Google Scholar] [PubMed]
- Musey, L.; Hughes, J.; Schacker, T.; Shea, T.; Corey, L.; McElrath, M.J. Cytotoxic-T-cell responses, viral load, and disease progression in early human immunodeficiency virus type 1 infection. N. Engl. J. Med. 1997, 337, 1267–1274. [Google Scholar] [CrossRef] [PubMed]
- Sengupta, S.; Siliciano, R.F. Targeting the Latent Reservoir for HIV-1. Immunity 2018, 48, 872–895. [Google Scholar] [CrossRef] [Green Version]
- Borrow, P.; Lewicki, H.; Wei, X.; Horwitz, M.S.; Peffer, N.; Meyers, H.; Nelson, J.A.; Gairin, J.E.; Hahn, B.H.; Oldstone, M.B.; et al. Antiviral pressure exerted by HIV-1-specific cytotoxic T lymphocytes (CTLs) during primary infection demonstrated by rapid selection of CTL escape virus. Nat. Med. 1997, 3, 205–211. [Google Scholar] [CrossRef]
- Goonetilleke, N.; Liu, M.K.; Salazar-Gonzalez, J.F.; Ferrari, G.; Giorgi, E.; Ganusov, V.V.; Keele, B.F.; Learn, G.H.; Turnbull, E.L.; Salazar, M.G.; et al. The first T cell response to transmitted/founder virus contributes to the control of acute viremia in HIV-1 infection. J. Exp. Med. 2009, 206, 1253–1272. [Google Scholar] [CrossRef]
- Price, D.A.; Goulder, P.J.; Klenerman, P.; Sewell, A.K.; Easterbrook, P.J.; Troop, M.; Bangham, C.R.; Phillips, R.E. Positive selection of HIV-1 cytotoxic T lymphocyte escape variants during primary infection. Proc. Natl. Acad. Sci. USA 1997, 94, 1890–1895. [Google Scholar] [CrossRef] [Green Version]
- Walker, B.; McMichael, A. The T-cell response to HIV. Cold Spring Harb. Perspect. Med. 2012, 2, a007054. [Google Scholar] [CrossRef] [Green Version]
- Deng, K.; Pertea, M.; Rongvaux, A.; Wang, L.; Durand, C.M.; Ghiaur, G.; Lai, J.; McHugh, H.L.; Hao, H.; Zhang, H.; et al. Broad CTL response is required to clear latent HIV-1 due to dominance of escape mutations. Nature 2015, 517, 381–385. [Google Scholar] [CrossRef] [Green Version]
- Smith, K.N.; Mailliard, R.B.; Piazza, P.A.; Fischer, W.; Korber, B.T.; Fecek, R.J.; Ratner, D.; Gupta, P.; Mullins, J.I.; Rinaldo, C.R. Effective Cytotoxic T Lymphocyte Targeting of Persistent HIV-1 during Antiretroviral Therapy Requires Priming of Naive CD8+ T Cells. Mbio 2016, 7, e00473-16, Erratum in: MBio. 2016;7(4). pii: e01012-16. doi: 10.1128/mBio.01012-16. [Google Scholar] [CrossRef] [Green Version]
- Qin, K.; Boppana, S.; Du, V.Y.; Carlson, J.M.; Yue, L.; Dilernia, D.A.; Hunter, E.; Mailliard, R.B.; Mallal, S.A.; Bansal, A.; et al. CD8 T cells targeting adapted epitopes in chronic HIV infection promote dendritic cell maturation and CD4 T cell trans-infection. PLoS Pathog. 2019, 15, e1007970. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Theiler, J.; Korber, B. Graph-based optimization of epitope coverage for vaccine antigen design. Stat. Med. 2018, 37, 181–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gaiha, G.D.; Rossin, E.J.; Urbach, J.; Landeros, C.; Collins, D.R.; Nwonu, C.; Muzhingi, I.; Anahtar, M.N.; Waring, O.M.; Piechocka-Trocha, A.; et al. Structural topology defines protective CD8+ T cell epitopes in the HIV proteome. Science 2019, 364, 480–484. [Google Scholar] [CrossRef] [PubMed]
- Bucks, C.M.; Norton, J.A.; Boesteanu, A.C.; Mueller, Y.M.; Katsikis, P.D. Chronic antigen stimulation alone is sufficient to drive CD8+ T cell exhaustion. J. Immunol. 2009, 182, 6697–6708. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cockerham, L.R.; Jain, V.; Sinclair, E.; Glidden, D.V.; Hartogenesis, W.; Hatano, H.; Hunt, P.W.; Martin, J.N.; Pilcher, C.D.; Sekaly, R.; et al. Programmed death-1 expression on CD4+ and CD8+ T cells in treated and untreated HIV disease. Aids 2014, 28, 1749–1758. [Google Scholar] [CrossRef] [Green Version]
- Day, C.L.; Kaufmann, D.E.; Kiepiela, P.; Brown, J.A.; Moodley, E.S.; Reddy, S.; Mackey, E.W.; Miller, J.D.; Leslie, A.J.; DePierres, C.; et al. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature 2006, 443, 350–354. [Google Scholar] [CrossRef]
- Streeck, H.; Brumme, Z.L.; Anastario, M.; Cohen, K.W.; Jolin, J.S.; Meier, A.; Brumme, C.J.; Rosenberg, E.S.; Alter, G.; Allen, T.M.; et al. Antigen load and viral sequence diversification determine the functional profile of HIV-1-specific CD8+ T cells. PLoS Med. 2008, 5, e100. [Google Scholar] [CrossRef]
- Kalams, S.A.; Buchbinder, S.P.; Rosenberg, E.S.; Billingsley, J.M.; Colbert, D.S.; Jones, N.G.; Shea, A.K.; Trocha, A.K.; Walker, B.D. Association between virus-specific cytotoxic T-lymphocyte and helper responses in human immunodeficiency virus type 1 infection. J. Virol. 1999, 73, 6715–6720. [Google Scholar]
- Kahan, S.M.; Wherry, E.J.; Zajac, A.J. T cell exhaustion during persistent viral infections. Virology 2015, 479, 180–193. [Google Scholar] [CrossRef]
- Trautmann, L.; Janbazian, L.; Chomont, N.; Said, E.A.; Gimmig, S.; Bessette, B.; Boulassel, M.R.; Delwart, E.; Sepulveda, H.; Balderas, R.S.; et al. Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nat. Med. 2006, 12, 1198–1202. [Google Scholar] [CrossRef]
- Petrovas, C.; Casazza, J.P.; Brenchley, J.M.; Price, D.A.; Gostick, E.; Adams, W.C.; Precopio, M.L.; Schacker, T.; Roederer, M.; Douek, D.C.; et al. PD-1 is a regulator of virus-specific CD8+ T cell survival in HIV infection. J. Exp. Med. 2006, 203, 2281–2292. [Google Scholar] [CrossRef] [PubMed]
- Wherry, E.J.; Ha, S.J.; Kaech, S.M.; Haining, W.N.; Sarkar, S.; Kalia, V.; Subramaniam, S.; Blattman, J.N.; Barber, D.L.; Ahmed, R. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 2007, 27, 670–684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jones, R.B.; Ndhlovu, L.C.; Barbour, J.D.; Sheth, P.M.; Jha, A.R.; Long, B.R.; Wong, J.C.; Satkunarajah, M.; Schweneker, M.; Chapman, J.M.; et al. Tim-3 expression defines a novel population of dysfunctional T cells with highly elevated frequencies in progressive HIV-1 infection. J. Exp. Med. 2008, 205, 2763–2779. [Google Scholar] [CrossRef] [PubMed]
- Blackburn, S.D.; Shin, H.; Haining, W.N.; Zou, T.; Workman, C.J.; Polley, A.; Betts, M.R.; Freeman, G.J.; Vignali, D.A.; Wherry, E.J. Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat. Immunol. 2009, 10, 29–37. [Google Scholar] [CrossRef] [Green Version]
- Wherry, E.J.; Kurachi, M. Molecular and cellular insights into T cell exhaustion. Nat. Rev. Immunol. 2015, 15, 486–499. [Google Scholar] [CrossRef]
- Petrovas, C.; Ferrando-Martinez, S.; Gerner, M.Y.; Casazza, J.P.; Pegu, A.; Deleage, C.; Cooper, A.; Hataye, J.; Andrews, S.; Ambrozak, D.; et al. Follicular CD8 T cells accumulate in HIV infection and can kill infected cells in vitro via bispecific antibodies. Sci. Transl. Med. 2017, 9, eaag2285. [Google Scholar] [CrossRef] [Green Version]
- Barber, D.L.; Wherry, E.J.; Masopust, D.; Zhu, B.; Allison, J.P.; Sharpe, A.H.; Freeman, G.J.; Ahmed, R. Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 2006, 439, 682–687. [Google Scholar] [CrossRef]
- Blackburn, S.D.; Shin, H.; Freeman, G.J.; Wherry, E.J. Selective expansion of a subset of exhausted CD8 T cells by alphaPD-L1 blockade. Proc. Natl. Acad. Sci. USA 2008, 105, 15016–15021. [Google Scholar] [CrossRef] [Green Version]
- Gay, C.L.; Bosch, R.J.; Ritz, J.; Hataye, J.M.; Aga, E.; Tressler, R.L.; Mason, S.W.; Hwang, C.K.; Grasela, D.M.; Ray, N.; et al. Clinical Trial of the Anti-PD-L1 Antibody BMS-936559 in HIV-1 Infected Participants on Suppressive Antiretroviral Therapy. J. Infect. Dis. 2017, 215, 1725–1733. [Google Scholar] [CrossRef] [Green Version]
- Chomont, N.; El-Far, M.; Ancuta, P.; Trautmann, L.; Procopio, F.A.; Yassine-Diab, B.; Boucher, G.; Boulassel, M.R.; Ghattas, G.; Brenchley, J.M.; et al. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat. Med. 2009, 15, 893–900. [Google Scholar] [CrossRef]
- Fromentin, R.; Bakeman, W.; Lawani, M.B.; Khoury, G.; Hartogensis, W.; DaFonseca, S.; Killian, M.; Epling, L.; Hoh, R.; Sinclair, E.; et al. CD4+ T Cells Expressing PD-1, TIGIT and LAG-3 Contribute to HIV Persistence during ART. PLoS Pathog. 2016, 12, e1005761. [Google Scholar] [CrossRef] [PubMed]
- Bruner, K.M.; Murray, A.J.; Pollack, R.A.; Soliman, M.G.; Laskey, S.B.; Capoferri, A.A.; Lai, J.; Strain, M.C.; Lada, S.M.; Hoh, R.; et al. Defective proviruses rapidly accumulate during acute HIV-1 infection. Nat. Med. 2016, 22, 1043–1049. [Google Scholar] [CrossRef] [Green Version]
- Pollack, R.A.; Jones, R.B.; Pertea, M.; Bruner, K.M.; Martin, A.R.; Thomas, A.S.; Capoferri, A.A.; Beg, S.A.; Huang, S.H.; Karandish, S.; et al. Defective HIV-1 Proviruses Are Expressed and Can Be Recognized by Cytotoxic T Lymphocytes, which Shape the Proviral Landscape. Cell Host Microbe 2017, 21, 494–506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ho, Y.C.; Shan, L.; Hosmane, N.N.; Wang, J.; Laskey, S.B.; Rosenbloom, D.I.; Lai, J.; Blankson, J.N.; Siliciano, J.D.; Siliciano, R.F. Replication-competent noninduced proviruses in the latent reservoir increase barrier to HIV-1 cure. Cell 2013, 155, 540–551. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Imamichi, H.; Dewar, R.L.; Adelsberger, J.W.; Rehm, C.A.; O’Doherty, U.; Paxinos, E.E.; Fauci, A.S.; Lane, H.C. Defective HIV-1 proviruses produce novel protein-coding RNA species in HIV-infected patients on combination antiretroviral therapy. Proc. Natl. Acad. Sci. USA 2016, 113, 8783–8788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, S.H.; Ren, Y.; Thomas, A.S.; Chan, D.; Mueller, S.; Ward, A.R.; Patel, S.; Bollard, C.M.; Cruz, C.R.; Karandish, S.; et al. Latent HIV reservoirs exhibit inherent resistance to elimination by CD8+ T cells. J. Clin. Investig. 2018, 128, 876–889. [Google Scholar] [CrossRef]
- Maldarelli, F.; Wu, X.; Su, L.; Simonetti, F.R.; Shao, W.; Hill, S.; Spindler, J.; Ferris, A.L.; Mellors, J.W.; Kearney, M.F.; et al. HIV latency. Specific HIV integration sites are linked to clonal expansion and persistence of infected cells. Science 2014, 345, 179–183. [Google Scholar] [CrossRef] [Green Version]
- Wagner, T.A.; McLaughlin, S.; Garg, K.; Cheung, C.Y.; Larsen, B.B.; Styrchak, S.; Huang, H.C.; Edlefsen, P.T.; Mullins, J.I.; Frenkel, L.M. HIV latency. Proliferation of cells with HIV integrated into cancer genes contributes to persistent infection. Science 2014, 345, 570–573. [Google Scholar] [CrossRef] [Green Version]
- Bui, J.K.; Sobolewski, M.D.; Keele, B.F.; Spindler, J.; Musick, A.; Wiegand, A.; Luke, B.T.; Shao, W.; Hughes, S.H.; Coffin, J.M.; et al. Proviruses with identical sequences comprise a large fraction of the replication-competent HIV reservoir. PLoS Pathog. 2017, 13, e1006283. [Google Scholar] [CrossRef]
- Hosmane, N.N.; Kwon, K.J.; Bruner, K.M.; Capoferri, A.A.; Beg, S.; Rosenbloom, D.I.; Keele, B.F.; Ho, Y.C.; Siliciano, J.D.; Siliciano, R.F. Proliferation of latently infected CD4+ T cells carrying replication-competent HIV-1: Potential role in latent reservoir dynamics. J. Exp. Med. 2017, 214, 959–972. [Google Scholar] [CrossRef]
- Cohn, L.B.; Silva, I.T.; Oliveira, T.Y.; Rosales, R.A.; Parrish, E.H.; Learn, G.H.; Hahn, B.H.; Czartoski, J.L.; McElrath, M.J.; Lehmann, C.; et al. HIV-1 integration landscape during latent and active infection. Cell 2015, 160, 420–432. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fukazawa, Y.; Lum, R.; Okoye, A.A.; Park, H.; Matsuda, K.; Bae, J.Y.; Hagen, S.I.; Shoemaker, R.; Deleage, C.; Lucero, C.; et al. B cell follicle sanctuary permits persistent productive simian immunodeficiency virus infection in elite controllers. Nat. Med. 2015, 21, 132–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Amet, T.; Son, Y.M.; Jiang, L.; Cheon, I.S.; Huang, S.; Gupta, S.K.; Dent, A.L.; Montaner, L.J.; Yu, Q.; Sun, J. BCL6 represses antiviral resistance in follicular T helper cells. J. Leukoc. Biol. 2017, 102, 527–536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kohler, S.L.; Pham, M.N.; Folkvord, J.M.; Arends, T.; Miller, S.M.; Miles, B.; Meditz, A.L.; McCarter, M.; Levy, D.N.; Connick, E. Germinal Center T Follicular Helper Cells Are Highly Permissive to HIV-1 and Alter Their Phenotype during Virus Replication. J. Immunol. 2016, 196, 2711–2722. [Google Scholar] [CrossRef] [Green Version]
- Miller, S.M.; Miles, B.; Guo, K.; Folkvord, J.; Meditz, A.L.; McCarter, M.D.; Levy, D.N.; MaWhinney, S.; Santiago, M.L.; Connick, E. Follicular Regulatory T Cells Are Highly Permissive to R5-Tropic HIV-1. J. Virol. 2017, 91, e00430-17. [Google Scholar] [CrossRef] [Green Version]
- Connick, E.; Mattila, T.; Folkvord, J.M.; Schlichtemeier, R.; Meditz, A.L.; Ray, M.G.; McCarter, M.D.; Mawhinney, S.; Hage, A.; White, C.; et al. CTL fail to accumulate at sites of HIV-1 replication in lymphoid tissue. J. Immunol. 2007, 178, 6975–6983. [Google Scholar] [CrossRef]
- Folkvord, J.M.; Armon, C.; Connick, E. Lymphoid follicles are sites of heightened human immunodeficiency virus type 1 (HIV-1) replication and reduced antiretroviral effector mechanisms. AIDS Res. Hum. Retrovir. 2005, 21, 363–370. [Google Scholar] [CrossRef]
- Perreau, M.; Savoye, A.L.; De Crignis, E.; Corpataux, J.M.; Cubas, R.; Haddad, E.K.; De Leval, L.; Graziosi, C.; Pantaleo, G. Follicular helper T cells serve as the major CD4 T cell compartment for HIV-1 infection, replication, and production. J. Exp. Med. 2013, 210, 143–156. [Google Scholar] [CrossRef]
- Hufert, F.T.; van Lunzen, J.; Janossy, G.; Bertram, S.; Schmitz, J.; Haller, O.; Racz, P.; von Laer, D. Germinal centre CD4+ T cells are an important site of HIV replication in vivo. Aids 1997, 11, 849–857. [Google Scholar] [CrossRef]
- Leong, Y.A.; Chen, Y.; Ong, H.S.; Wu, D.; Man, K.; Deleage, C.; Minnich, M.; Meckiff, B.J.; Wei, Y.; Hou, Z.; et al. CXCR5+ follicular cytotoxic T cells control viral infection in B cell follicles. Nat. Immunol. 2016, 17, 1187–1196. [Google Scholar] [CrossRef]
- Im, S.J.; Hashimoto, M.; Gerner, M.Y.; Lee, J.; Kissick, H.T.; Burger, M.C.; Shan, Q.; Hale, J.S.; Lee, J.; Nasti, T.H.; et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 2016, 537, 417–421. [Google Scholar] [CrossRef] [PubMed]
- Quigley, M.F.; Gonzalez, V.D.; Granath, A.; Andersson, J.; Sandberg, J.K. CXCR5+ CCR7− CD8 T cells are early effector memory cells that infiltrate tonsil B cell follicles. Eur. J. Immunol. 2007, 37, 3352–3362. [Google Scholar] [CrossRef] [PubMed]
- Bronnimann, M.P.; Skinner, P.J.; Connick, E. The B-Cell Follicle in HIV Infection: Barrier to a Cure. Front. Immunol. 2018, 9, 20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, S.; Folkvord, J.M.; Rakasz, E.G.; Abdelaal, H.M.; Wagstaff, R.K.; Kovacs, K.J.; Kim, H.O.; Sawahata, R.; MaWhinney, S.; Masopust, D.; et al. Simian Immunodeficiency Virus-Producing Cells in Follicles Are Partially Suppressed by CD8+ Cells In Vivo. J. Virol. 2016, 90, 11168–11180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ayala, V.I.; Deleage, C.; Trivett, M.T.; Jain, S.; Coren, L.V.; Breed, M.W.; Kramer, J.A.; Thomas, J.A.; Estes, J.D.; Lifson, J.D.; et al. CXCR5-Dependent Entry of CD8 T Cells into Rhesus Macaque B-Cell Follicles Achieved through T-Cell Engineering. J. Virol. 2017, 91, e02507-16. [Google Scholar] [CrossRef] [Green Version]
- Mylvaganam, G.H.; Rios, D.; Abdelaal, H.M.; Iyer, S.; Tharp, G.; Mavigner, M.; Hicks, S.; Chahroudi, A.; Ahmed, R.; Bosinger, S.E.; et al. Dynamics of SIV-specific CXCR5+ CD8 T cells during chronic SIV infection. Proc. Natl. Acad. Sci. USA 2017, 114, 1976–1981. [Google Scholar] [CrossRef] [Green Version]
- Pegu, A.; Asokan, M.; Wu, L.; Wang, K.; Hataye, J.; Casazza, J.P.; Guo, X.; Shi, W.; Georgiev, I.; Zhou, T.; et al. Activation and lysis of human CD4 cells latently infected with HIV-1. Nat. Commun. 2015, 6, 8447. [Google Scholar] [CrossRef]
- Deeks, S.G. HIV: Shock and kill. Nature 2012, 487, 439–440. [Google Scholar] [CrossRef]
- Shan, L.; Deng, K.; Gao, H.; Xing, S.; Capoferri, A.A.; Durand, C.M.; Rabi, S.A.; Laird, G.M.; Kim, M.; Hosmane, N.N.; et al. Transcriptional Reprogramming during Effector-to-Memory Transition Renders CD4+ T Cells Permissive for Latent HIV-1 Infection. Immunity 2017, 47, 766–775. [Google Scholar] [CrossRef] [Green Version]
- Kim, Y.; Anderson, J.L.; Lewin, S.R. Getting the “Kill” into “Shock and Kill”: Strategies to Eliminate Latent HIV. Cell Host Microbe 2018, 23, 14–26. [Google Scholar] [CrossRef]
- Spivak, A.M.; Planelles, V. Novel Latency Reversal Agents for HIV-1 Cure. Annu. Rev. Med. 2018, 69, 421–436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rasmussen, T.A.; Tolstrup, M.; Moller, H.J.; Brinkmann, C.R.; Olesen, R.; Erikstrup, C.; Laursen, A.L.; Ostergaard, L.; Sogaard, O.S. Activation of latent human immunodeficiency virus by the histone deacetylase inhibitor panobinostat: A pilot study to assess effects on the central nervous system. Open Forum Infect. Dis. 2015, 2, ofv037. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kulkosky, J.; Sullivan, J.; Xu, Y.; Souder, E.; Hamer, D.H.; Pomerantz, R.J. Expression of latent HAART-persistent HIV type 1 induced by novel cellular activating agents. AIDS Res. Hum. Retrovir. 2004, 20, 497–505. [Google Scholar] [CrossRef] [PubMed]
- Williams, S.A.; Chen, L.F.; Kwon, H.; Fenard, D.; Bisgrove, D.; Verdin, E.; Greene, W.C. Prostratin antagonizes HIV latency by activating NF-kappaB. J. Biol. Chem. 2004, 279, 42008–42017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bullen, C.K.; Laird, G.M.; Durand, C.M.; Siliciano, J.D.; Siliciano, R.F. New ex vivo approaches distinguish effective and ineffective single agents for reversing HIV-1 latency in vivo. Nat. Med. 2014, 20, 425–429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Clutton, G.; Xu, Y.; Baldoni, P.L.; Mollan, K.R.; Kirchherr, J.; Newhard, W.; Cox, K.; Kuruc, J.D.; Kashuba, A.; Barnard, R.; et al. The differential short- and long-term effects of HIV-1 latency-reversing agents on T cell function. Sci. Rep. 2016, 6, 30749. [Google Scholar] [CrossRef]
- Lam, A.P.; Sparano, J.A.; Vinciguerra, V.; Ocean, A.J.; Christos, P.; Hochster, H.; Camacho, F.; Goel, S.; Mani, S.; Kaubisch, A. Phase II study of paclitaxel plus the protein kinase C inhibitor bryostatin-1 in advanced pancreatic carcinoma. Am. J. Clin. Oncol. 2010, 33, 121–124. [Google Scholar] [CrossRef] [Green Version]
- Smith, B.D.; Jones, R.J.; Cho, E.; Kowalski, J.; Karp, J.E.; Gore, S.D.; Vala, M.; Meade, B.; Baker, S.D.; Zhao, M.; et al. Differentiation therapy in poor risk myeloid malignancies: Results of a dose finding study of the combination bryostatin-1 and GM-CSF. Leuk. Res. 2011, 35, 87–94. [Google Scholar] [CrossRef] [Green Version]
- Morgan, R.J., Jr.; Leong, L.; Chow, W.; Gandara, D.; Frankel, P.; Garcia, A.; Lenz, H.J.; Doroshow, J.H. Phase II trial of bryostatin-1 in combination with cisplatin in patients with recurrent or persistent epithelial ovarian cancer: A California cancer consortium study. Investig. New Drugs 2012, 30, 723–728. [Google Scholar] [CrossRef] [Green Version]
- Ajani, J.A.; Jiang, Y.; Faust, J.; Chang, B.B.; Ho, L.; Yao, J.C.; Rousey, S.; Dakhil, S.; Cherny, R.C.; Craig, C.; et al. A multi-center phase II study of sequential paclitaxel and bryostatin-1 (NSC 339555) in patients with untreated, advanced gastric or gastroesophageal junction adenocarcinoma. Investig. New Drugs 2006, 24, 353–357. [Google Scholar] [CrossRef] [PubMed]
- Laird, G.M.; Bullen, C.K.; Rosenbloom, D.I.; Martin, A.R.; Hill, A.L.; Durand, C.M.; Siliciano, J.D.; Siliciano, R.F. Ex vivo analysis identifies effective HIV-1 latency-reversing drug combinations. J. Clin. Investig. 2015, 125, 1901–1912. [Google Scholar] [CrossRef] [PubMed]
- Cillo, A.R.; Sobolewski, M.D.; Bosch, R.J.; Fyne, E.; Piatak, M., Jr.; Coffin, J.M.; Mellors, J.W. Quantification of HIV-1 latency reversal in resting CD4+ T cells from patients on suppressive antiretroviral therapy. Proc. Natl. Acad. Sci. USA 2014, 111, 7078–7083. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Walker-Sperling, V.E.; Pohlmeyer, C.W.; Tarwater, P.M.; Blankson, J.N. The Effect of Latency Reversal Agents on Primary CD8+ T Cells: Implications for Shock and Kill Strategies for Human Immunodeficiency Virus Eradication. EBioMedicine 2016, 8, 217–229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pace, M.; Williams, J.; Kurioka, A.; Gerry, A.B.; Jakobsen, B.; Klenerman, P.; Nwokolo, N.; Fox, J.; Fidler, S.; Frater, J.; et al. Histone Deacetylase Inhibitors Enhance CD4 T Cell Susceptibility to NK Cell Killing but Reduce NK Cell Function. PLoS Pathog. 2016, 12, e1005782. [Google Scholar] [CrossRef] [Green Version]
- Jones, R.B.; O’Connor, R.; Mueller, S.; Foley, M.; Szeto, G.L.; Karel, D.; Lichterfeld, M.; Kovacs, C.; Ostrowski, M.A.; Trocha, A.; et al. Histone deacetylase inhibitors impair the elimination of HIV-infected cells by cytotoxic T-lymphocytes. PLoS Pathog. 2014, 10, e1004287. [Google Scholar] [CrossRef] [Green Version]
- Tsai, A.; Irrinki, A.; Kaur, J.; Cihlar, T.; Kukolj, G.; Sloan, D.D.; Murry, J.P. Toll-Like Receptor 7 Agonist GS-9620 Induces HIV Expression and HIV-Specific Immunity in Cells from HIV-Infected Individuals on Suppressive Antiretroviral Therapy. J. Virol. 2017, 91, e02166-16. [Google Scholar] [CrossRef] [Green Version]
- Borducchi, E.N.; Cabral, C.; Stephenson, K.E.; Liu, J.; Abbink, P.; Ng’ang’a, D.; Nkolola, J.P.; Brinkman, A.L.; Peter, L.; Lee, B.C.; et al. Ad26/MVA therapeutic vaccination with TLR7 stimulation in SIV-infected rhesus monkeys. Nature 2016, 540, 284. [Google Scholar] [CrossRef]
- Borducchi, E.N.; Liu, J.; Nkolola, J.P.; Cadena, A.M.; Yu, W.H.; Fischinger, S.; Broge, T.; Abbink, P.; Mercado, N.B.; Chandrashekar, A.; et al. Publisher Correction: Antibody and TLR7 agonist delay viral rebound in SHIV-infected monkeys. Nature 2018, 564, E8. [Google Scholar] [CrossRef]
- Vibholm, L.; Schleimann, M.H.; Hojen, J.F.; Benfield, T.; Offersen, R.; Rasmussen, K.; Olesen, R.; Dige, A.; Agnholt, J.; Grau, J.; et al. Short-Course Toll-Like Receptor 9 Agonist Treatment Impacts Innate Immunity and Plasma Viremia in Individuals With Human Immunodeficiency Virus Infection. Clin. Infect. Dis. 2017, 64, 1686–1695. [Google Scholar] [CrossRef]
- Jones, R.B.; Mueller, S.; O’Connor, R.; Rimpel, K.; Sloan, D.D.; Karel, D.; Wong, H.C.; Jeng, E.K.; Thomas, A.S.; Whitney, J.B.; et al. A Subset of Latency-Reversing Agents Expose HIV-Infected Resting CD4+ T-Cells to Recognition by Cytotoxic T-Lymphocytes. PLoS Pathog. 2016, 12, e1005545. [Google Scholar] [CrossRef] [Green Version]
- Liu, K.; Catalfamo, M.; Li, Y.; Henkart, P.A.; Weng, N.P. IL-15 mimics T cell receptor crosslinking in the induction of cellular proliferation, gene expression, and cytotoxicity in CD8+ memory T cells. Proc. Natl. Acad. Sci. USA 2002, 99, 6192–6197. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lucas, M.; Schachterle, W.; Oberle, K.; Aichele, P.; Diefenbach, A. Dendritic cells prime natural killer cells by trans-presenting interleukin 15. Immunity 2007, 26, 503–517. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mortier, E.; Woo, T.; Advincula, R.; Gozalo, S.; Ma, A. IL-15Ralpha chaperones IL-15 to stable dendritic cell membrane complexes that activate NK cells via trans presentation. J. Exp. Med. 2008, 205, 1213–1225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Waldmann, T.A. The biology of interleukin-2 and interleukin-15: Implications for cancer therapy and vaccine design. Nat. Rev. Immunol. 2006, 6, 595–601. [Google Scholar] [CrossRef] [PubMed]
- Lu, W.; Wu, X.; Lu, Y.; Guo, W.; Andrieu, J.M. Therapeutic dendritic-cell vaccine for simian AIDS. Nat. Med. 2003, 9, 27–32. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lu, W.; Arraes, L.C.; Ferreira, W.T.; Andrieu, J.M. Therapeutic dendritic-cell vaccine for chronic HIV-1 infection. Nat. Med. 2004, 10, 1359–1365. [Google Scholar] [CrossRef]
- Connolly, N.C.; Whiteside, T.L.; Wilson, C.; Kondragunta, V.; Rinaldo, C.R.; Riddler, S.A. Therapeutic immunization with human immunodeficiency virus type 1 (HIV-1) peptide-loaded dendritic cells is safe and induces immunogenicity in HIV-1-infected individuals. Clin. Vaccine Immunol. 2008, 15, 284–292. [Google Scholar] [CrossRef] [Green Version]
- Garcia, F.; Climent, N.; Guardo, A.C.; Gil, C.; Leon, A.; Autran, B.; Lifson, J.D.; Martinez-Picado, J.; Dalmau, J.; Clotet, B.; et al. A dendritic cell-based vaccine elicits T cell responses associated with control of HIV-1 replication. Sci. Transl. Med. 2013, 5, 166ra2. [Google Scholar] [CrossRef]
- Jacobson, J.M.; Routy, J.P.; Welles, S.; DeBenedette, M.; Tcherepanova, I.; Angel, J.B.; Asmuth, D.M.; Stein, D.K.; Baril, J.G.; McKellar, M.; et al. Dendritic Cell Immunotherapy for HIV-1 Infection Using Autologous HIV-1 RNA: A Randomized, Double-Blind, Placebo-Controlled Clinical Trial. J. Acquir. Immune Defic. Syndr. 2016, 72, 31. [Google Scholar] [CrossRef] [Green Version]
- Gay, C.L.; DeBenedette, M.A.; Tcherepanova, I.Y.; Gamble, A.; Lewis, W.E.; Cope, A.B.; Kuruc, J.D.; McGee, K.S.; Kearney, M.F.; Coffin, J.M.; et al. Immunogenicity of AGS-004 Dendritic Cell Therapy in Patients Treated During Acute HIV Infection. AIDS Res. Hum. Retrovir. 2018, 34, 111–122. [Google Scholar] [CrossRef]
- Routy, J.P.; Nicolette, C. Arcelis AGS-004 dendritic cell-based immunotherapy for HIV infection. Immunotherapy 2010, 2, 467–476. [Google Scholar] [CrossRef] [PubMed]
- Routy, J.P.; Boulassel, M.R.; Yassine-Diab, B.; Nicolette, C.; Healey, D.; Jain, R.; Landry, C.; Yegorov, O.; Tcherepanova, I.; Monesmith, T.; et al. Immunologic activity and safety of autologous HIV RNA-electroporated dendritic cells in HIV-1 infected patients receiving antiretroviral therapy. Clin. Immunol. 2010, 134, 140–147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Guardo, A.C.; Joe, P.T.; Miralles, L.; Bargalló, M.E.; Mothe, B.; Krasniqi, A.; Heirman, C.; García, F.; Thielemans, K.; Brander, C.; et al. Preclinical evaluation of an mRNA HIV vaccine combining rationally selected antigenic sequences and adjuvant signals (HTI-TriMix). Aids 2017, 31, 321–332. [Google Scholar] [CrossRef] [PubMed]
- Bonehill, A.; Tuyaerts, S.; Van Nuffel, A.M.; Heirman, C.; Bos, T.J.; Fostier, K.; Neyns, B.; Thielemans, K. Enhancing the T-cell stimulatory capacity of human dendritic cells by co-electroporation with CD40L, CD70 and constitutively active TLR4 encoding mRNA. Mol. Ther. 2008, 16, 1170–1180. [Google Scholar] [CrossRef]
- Mothe, B.; Hu, X.; Llano, A.; Rosati, M.; Olvera, A.; Kulkarni, V.; Valentin, A.; Alicea, C.; Pilkington, G.R.; Sardesai, N.Y.; et al. A human immune data-informed vaccine concept elicits strong and broad T-cell specificities associated with HIV-1 control in mice and macaques. J. Transl. Med. 2015, 13, 60. [Google Scholar] [CrossRef] [Green Version]
- Ahmad, A.; Rinaldo, C.R. A novel anti-HIV immunotherapy to cure HIV. Aids 2017, 31, 447–449. [Google Scholar] [CrossRef]
- Surenaud, M.; Montes, M.; Arlehamn, C.S.L.; Sette, A.; Banchereau, J.; Palucka, K.; Lelievre, J.D.; Lacabaratz, C.; Levy, Y. Anti-HIV potency of T-cell responses elicited by dendritic cell therapeutic vaccination. PLoS Pathog. 2019, 15, e1008011. [Google Scholar] [CrossRef]
- Coelho, A.V.; de Moura, R.R.; Kamada, A.J.; da Silva, R.C.; Guimaraes, R.L.; Brandao, L.A.; de Alencar, L.C.; Crovella, S. Dendritic Cell-Based Immunotherapies to Fight HIV: How Far from a Success Story? A Systematic Review and Meta-Analysis. Int. J. Mol. Sci. 2016, 17, 1985. [Google Scholar] [CrossRef] [Green Version]
- Garcia, F.; Routy, J.P. Challenges in dendritic cells-based therapeutic vaccination in HIV-1 infection Workshop in dendritic cell-based vaccine clinical trials in HIV-1. Vaccine 2011, 29, 6454–6463. [Google Scholar] [CrossRef]
- Brezar, V.; Ruffin, N.; Richert, L.; Surenaud, M.; Lacabaratz, C.; Palucka, K.; Thiebaut, R.; Banchereau, J.; Levy, Y.; Seddiki, N. Decreased HIV-specific T-regulatory responses are associated with effective DC-vaccine induced immunity. PLoS Pathog. 2015, 11, e1004752. [Google Scholar] [CrossRef]
- Levy, Y.; Thiebaut, R.; Montes, M.; Lacabaratz, C.; Sloan, L.; King, B.; Perusat, S.; Harrod, C.; Cobb, A.; Roberts, L.K.; et al. Dendritic cell-based therapeutic vaccine elicits polyfunctional HIV-specific T-cell immunity associated with control of viral load. Eur. J. Immunol. 2014, 44, 2802–2810. [Google Scholar] [CrossRef] [PubMed]
- Izquierdo-Useros, N.; Lorizate, M.; McLaren, P.J.; Telenti, A.; Krausslich, H.G.; Martinez-Picado, J. HIV-1 capture and transmission by dendritic cells: The role of viral glycolipids and the cellular receptor Siglec-1. PLoS Pathog. 2014, 10, e1004146. [Google Scholar] [CrossRef] [PubMed]
- Rappocciolo, G.; Jais, M.; Piazza, P.; Reinhart, T.A.; Berendam, S.J.; Garcia-Exposito, L.; Gupta, P.; Rinaldo, C.R. Alterations in cholesterol metabolism restrict HIV-1 trans infection in nonprogressors. MBio 2014, 5, e01031-13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kumar, N.A.; van der Sluis, R.M.; Mota, T.; Pascoe, R.; Evans, V.A.; Lewin, S.R.; Cameron, P.U. Myeloid Dendritic Cells Induce HIV Latency in Proliferating CD4+ T Cells. J. Immunol. 2018, 201, 1468–1477. [Google Scholar] [CrossRef] [Green Version]
- Evans, V.A.; Kumar, N.; Filali, A.; Procopio, F.A.; Yegorov, O.; Goulet, J.P.; Saleh, S.; Haddad, E.K.; da Fonseca Pereira, C.; Ellenberg, P.C.; et al. Myeloid dendritic cells induce HIV-1 latency in non-proliferating CD4+ T cells. PLoS Pathog. 2013, 9, e1003799. [Google Scholar] [CrossRef] [Green Version]
- Zhao, X.Q.; Huang, X.L.; Gupta, P.; Borowski, L.; Fan, Z.; Watkins, S.C.; Thomas, E.K.; Rinaldo, C.R., Jr. Induction of anti-human immunodeficiency virus type 1 (HIV-1) CD8+ and CD4+ T-cell reactivity by dendritic cells loaded with HIV-1 X4-infected apoptotic cells. J. Virol. 2002, 76, 3007–3014. [Google Scholar] [CrossRef] [Green Version]
- Macatangay, B.J.; Riddler, S.A.; Wheeler, N.D.; Spindler, J.; Lawani, M.; Hong, F.; Buffo, M.J.; Whiteside, T.L.; Kearney, M.F.; Mellors, J.W.; et al. Therapeutic Vaccination With Dendritic Cells Loaded With Autologous HIV Type 1-Infected Apoptotic Cells. J. Infect. Dis. 2016, 213, 1400–1409. [Google Scholar] [CrossRef] [Green Version]
- Van der Sluis, R.M.; van Montfort, T.; Pollakis, G.; Sanders, R.W.; Speijer, D.; Berkhout, B.; Jeeninga, R.E. Dendritic cell-induced activation of latent HIV-1 provirus in actively proliferating primary T lymphocytes. PLoS Pathog. 2013, 9, e1003259. [Google Scholar] [CrossRef] [Green Version]
- Marini, A.; Harper, J.M.; Romerio, F. An In Vitro System to Model the Establishment and Reactivation of HIV-1 Latency. J. Immunol. 2008, 181, 7713–7720. [Google Scholar] [CrossRef]
- Ren, X.X.; Ma, L.; Sun, W.W.; Kuang, W.D.; Li, T.S.; Jin, X.; Wang, J.H. Dendritic cells maturated by co-culturing with HIV-1 latently infected Jurkat T cells or stimulating with AIDS-associated pathogens secrete TNF-alpha to reactivate HIV-1 from latency. Virulence 2017, 8, 1732–1743. [Google Scholar] [CrossRef] [Green Version]
- Sanyal, A.; Mailliard, R.B.; Rinaldo, C.R.; Ratner, D.; Ding, M.; Chen, Y.; Zerbato, J.M.; Giacobbi, N.S.; Venkatachari, N.J.; Patterson, B.K.; et al. Novel assay reveals a large, inducible, replication-competent HIV-1 reservoir in resting CD4+ T cells. Nat. Med. 2017, 23, 885–889. [Google Scholar] [CrossRef] [PubMed]
- Spina, C.A.; Anderson, J.; Archin, N.M.; Bosque, A.; Chan, J.; Famiglietti, M.; Greene, W.C.; Kashuba, A.; Lewin, S.R.; Margolis, D.M.; et al. An in-depth comparison of latent HIV-1 reactivation in multiple cell model systems and resting CD4+ T cells from aviremic patients. PLoS Pathog. 2013, 9, e1003834. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zerbato, J.M.; Serrao, E.; Lenzi, G.; Kim, B.; Ambrose, Z.; Watkins, S.C.; Engelman, A.N.; Sluis-Cremer, N. Establishment and Reversal of HIV-1 Latency in Naive and Central Memory CD4+ T Cells In Vitro. J. Virol. 2016, 90, 8059–8073. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marsden, M.D.; Loy, B.A.; Wu, X.; Ramirez, C.M.; Schrier, A.J.; Murray, D.; Shimizu, A.; Ryckbosch, S.M.; Near, K.E.; Chun, T.W.; et al. In vivo activation of latent HIV with a synthetic bryostatin analog effects both latent cell “kick” and “kill” in strategy for virus eradication. PLoS Pathog. 2017, 13, e1006575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Norton, T.D.; Miller, E.A.; Bhardwaj, N.; Landau, N.R. Vpx-containing dendritic cell vaccine induces CTLs and reactivates latent HIV-1 in vitro. Gene Ther. 2015, 22, 227. [Google Scholar] [CrossRef] [Green Version]
- Van Montfort, T.; van der Sluis, R.; Darcis, G.; Beaty, D.; Groen, K.; Pasternak, A.O.; Pollakis, G.; Vink, M.; Westerhout, E.M.; Hamdi, M.; et al. Dendritic cells potently purge latent HIV-1 beyond TCR-stimulation, activating the PI3K-Akt-mTOR pathway. Ebiomedicine 2019, 42, 97–108. [Google Scholar] [CrossRef] [Green Version]
- Van der Sluis, R.M.; van Capel, T.M.; Speijer, D.; Sanders, R.W.; Berkhout, B.; de Jong, E.C.; Jeeninga, R.E.; van Montfort, T. Dendritic cell type-specific HIV-1 activation in effector T cells: Implications for latent HIV-1 reservoir establishment. Aids 2015, 29, 1003–1014. [Google Scholar] [CrossRef]
- Douek, D.C.; Brenchley, J.M.; Betts, M.R.; Ambrozak, D.R.; Hill, B.J.; Okamoto, Y.; Casazza, J.P.; Kuruppu, J.; Kunstman, K.; Wolinsky, S.; et al. HIV preferentially infects HIV-specific CD4+ T cells. Nature 2002, 417, 95–98. [Google Scholar] [CrossRef]
- Brenchley, J.M.; Ruff, L.E.; Casazza, J.P.; Koup, R.A.; Price, D.A.; Douek, D.C. Preferential infection shortens the life span of human immunodeficiency virus-specific CD4+ T cells in vivo. J. Virol. 2006, 80, 6801–6809. [Google Scholar] [CrossRef] [Green Version]
- Demoustier, A.; Gubler, B.; Lambotte, O.; de Goer, M.G.; Wallon, C.; Goujard, C.; Delfraissy, J.F.; Taoufik, Y. In patients on prolonged HAART, a significant pool of HIV infected CD4 T cells are HIV-specific. Aids 2002, 16, 1749–1754. [Google Scholar] [CrossRef] [Green Version]
- Casazza, J.P.; Brenchley, J.M.; Hill, B.J.; Ayana, R.; Ambrozak, D.; Roederer, M.; Douek, D.C.; Betts, M.R.; Koup, R.A. Autocrine production of beta-chemokines protects CMV-Specific CD4 T cells from HIV infection. PLoS Pathog. 2009, 5, e1000646. [Google Scholar] [CrossRef] [PubMed]
- Stone, S.F.; Price, P.; French, M.A. Cytomegalovirus (CMV)-specific CD8+ T cells in individuals with HIV infection: Correlation with protection from CMV disease. J. Antimicrob. Chemother. 2006, 57, 585–588. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sylwester, A.W.; Mitchell, B.L.; Edgar, J.B.; Taormina, C.; Pelte, C.; Ruchti, F.; Sleath, P.R.; Grabstein, K.H.; Hosken, N.A.; Kern, F.; et al. Broadly targeted human cytomegalovirus-specific CD4+ and CD8+ T cells dominate the memory compartments of exposed subjects. J. Exp. Med. 2005, 202, 673–685. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abana, C.O.; Pilkinton, M.A.; Gaudieri, S.; Chopra, A.; McDonnell, W.J.; Wanjalla, C.; Barnett, L.; Gangula, R.; Hager, C.; Jung, D.K.; et al. Cytomegalovirus (CMV) Epitope-Specific CD4+ T Cells Are Inflated in HIV+ CMV+ Subjects. J. Immunol. 2017, 199, 3187–3201. [Google Scholar] [CrossRef] [PubMed]
- Komanduri, K.V.; Donahoe, S.M.; Moretto, W.J.; Schmidt, D.K.; Gillespie, G.; Ogg, G.S.; Roederer, M.; Nixon, D.F.; McCune, J.M. Direct measurement of CD4+ and CD8+ T-cell responses to CMV in HIV-1-infected subjects. Virology 2001, 279, 459–470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Naeger, D.M.; Martin, J.N.; Sinclair, E.; Hunt, P.W.; Bangsberg, D.R.; Hecht, F.; Hsue, P.; McCune, J.M.; Deeks, S.G. Cytomegalovirus-specific T cells persist at very high levels during long-term antiretroviral treatment of HIV disease. PLoS ONE 2010, 5, e8886. [Google Scholar] [CrossRef] [Green Version]
- Soderberg-Naucler, C. Treatment of cytomegalovirus infections beyond acute disease to improve human health. Expert Rev. Anti Infect. Ther. 2014, 12, 211–222. [Google Scholar] [CrossRef]
- Maidji, E.; Somsouk, M.; Rivera, J.M.; Hunt, P.W.; Stoddart, C.A. Replication of CMV in the gut of HIV-infected individuals and epithelial barrier dysfunction. PLoS Pathog. 2017, 13, e1006202. [Google Scholar] [CrossRef] [Green Version]
- Gianella, S.; Chaillon, A.; Mutlu, E.A.; Engen, P.A.; Voigt, R.M.; Keshavarzian, A.; Losurdo, J.; Chakradeo, P.; Lada, S.M.; Nakazawa, M.; et al. Effect of cytomegalovirus and Epstein-Barr virus replication on intestinal mucosal gene expression and microbiome composition of HIV-infected and uninfected individuals. Aids 2017, 31, 2059–2067. [Google Scholar] [CrossRef]
- Christensen-Quick, A.; Vanpouille, C.; Lisco, A.; Gianella, S. Cytomegalovirus and HIV Persistence: Pouring Gas on the Fire. Aids Res. Hum. Retrovir. 2017, 33, S-23. [Google Scholar] [CrossRef]
- Gianella, S.; Anderson, C.M.; Var, S.R.; Oliveira, M.F.; Lada, S.M.; Vargas, M.V.; Massanella, M.; Little, S.J.; Richman, D.D.; Strain, M.C.; et al. Replication of Human Herpesviruses Is Associated with Higher HIV DNA Levels during Antiretroviral Therapy Started at Early Phases of HIV Infection. J. Virol. 2016, 90, 3944–3952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Freeman, M.L.; Lederman, M.M.; Gianella, S. Partners in Crime: The Role of CMV in Immune Dysregulation and Clinical Outcome During HIV Infection. Curr. HIV/AIDS Rep. 2016, 13, 10–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Smith, D.M.; Nakazawa, M.; Freeman, M.L.; Anderson, C.M.; Oliveira, M.F.; Little, S.J.; Gianella, S. Asymptomatic CMV Replication During Early Human Immunodeficiency Virus (HIV) Infection Is Associated With Lower CD4/CD8 Ratio During HIV Treatment. Clin. Infect. Dis. 2016, 63, 1517–1524. [Google Scholar] [CrossRef] [PubMed]
- Poizot-Martin, I.; Allavena, C.; Duvivier, C.; Cano, C.E.; Guillouet de Salvador, F.; Rey, D.; Dellamonica, P.; Cuzin, L.; Cheret, A.; Hoen, B.; et al. CMV+ Serostatus Associates Negatively with CD4:CD8 Ratio Normalization in Controlled HIV-Infected Patients on cART. PLoS ONE 2016, 11, e0165774. [Google Scholar] [CrossRef]
- Dan, J.M.; Massanella, M.; Smith, D.M.; Spina, C.A.; Schrier, R.; Daar, E.S.; Dube, M.P.; Morris, S.R.; Gianella, S. Brief Report: Effect of CMV and HIV Transcription on CD57 and PD-1 T-Cell Expression During Suppressive ART. J. Acquir. Immune Defic. Syndr. 2016, 72, 133. [Google Scholar] [CrossRef]
- Agudelo-Hernandez, A.; Chen, Y.; Bullotta, A.; Buchanan, W.G.; Klamar-Blain, C.R.; Borowski, L.; Riddler, S.A.; Rinaldo, C.R.; Macatangay, B.J.C. Subclinical herpesvirus shedding among HIV-1-infected men on antiretroviral therapy. Aids 2017, 31, 2085–2094. [Google Scholar] [CrossRef]
- Johnson, E.L.; Howard, C.L.; Thurman, J.; Pontiff, K.; Johnson, E.S.; Chakraborty, R. Cytomegalovirus upregulates expression of CCR5 in central memory cord blood mononuclear cells, which may facilitate in utero HIV type 1 transmission. J. Infect. Dis. 2015, 211, 187–196. [Google Scholar] [CrossRef] [Green Version]
- Barry, P.A.; Pratt-Lowe, E.; Peterlin, B.M.; Luciw, P.A. Cytomegalovirus activates transcription directed by the long terminal repeat of human immunodeficiency virus type 1. J. Virol. 1990, 64, 2932–2940. [Google Scholar]
- McCarthy, M.; Auger, D.; He, J.; Wood, C. Cytomegalovirus and human herpesvirus-6 trans-activate the HIV-1 long terminal repeat via multiple response regions in human fetal astrocytes. J. Neurovirol. 1998, 4, 495–511. [Google Scholar] [CrossRef]
- Murayama, T.; Ohara, Y.; Obuchi, M.; Khabar, K.S.; Higashi, H.; Mukaida, N.; Matsushima, K. Human cytomegalovirus induces interleukin-8 production by a human monocytic cell line, THP-1, through acting concurrently on AP-1- and NF-kappaB-binding sites of the interleukin-8 gene. J. Virol. 1997, 71, 5692–5695. [Google Scholar]
- Saleh, S.; Lu, H.K.; Evans, V.; Harisson, D.; Zhou, J.; Jaworowski, A.; Sallmann, G.; Cheong, K.Y.; Mota, T.M.; Tennakoon, S.; et al. HIV integration and the establishment of latency in CCL19-treated resting CD4+ T cells require activation of NF-kappaB. Retrovirology 2016, 13, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gianella, S.; Anderson, C.M.; Vargas, M.V.; Richman, D.D.; Little, S.J.; Morris, S.R.; Smith, D.M. Cytomegalovirus DNA in semen and blood is associated with higher levels of proviral HIV DNA. J. Infect. Dis. 2013, 207, 898–902. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gianella, S.; Massanella, M.; Richman, D.D.; Little, S.J.; Spina, C.A.; Vargas, M.V.; Lada, S.M.; Daar, E.S.; Dube, M.P.; Haubrich, R.H.; et al. Cytomegalovirus replication in semen is associated with higher levels of proviral HIV DNA and CD4+ T cell activation during antiretroviral treatment. J. Virol. 2014, 88, 7818–7827. [Google Scholar] [CrossRef] [Green Version]
- Wagner, T.A.; McKernan, J.L.; Tobin, N.H.; Tapia, K.A.; Mullins, J.I.; Frenkel, L.M. An increasing proportion of monotypic HIV-1 DNA sequences during antiretroviral treatment suggests proliferation of HIV-infected cells. J. Virol. 2013, 87, 1770–1778. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Von Stockenstrom, S.; Odevall, L.; Lee, E.; Sinclair, E.; Bacchetti, P.; Killian, M.; Epling, L.; Shao, W.; Hoh, R.; Ho, T.; et al. Longitudinal Genetic Characterization Reveals That Cell Proliferation Maintains a Persistent HIV Type 1 DNA Pool During Effective HIV Therapy. J. Infect. Dis. 2015, 212, 596–607. [Google Scholar] [CrossRef] [PubMed]
- Henrich, T.J.; Hobbs, K.S.; Hanhauser, E.; Scully, E.; Hogan, L.E.; Robles, Y.P.; Leadabrand, K.S.; Marty, F.M.; Palmer, C.D.; Jost, S.; et al. Human Immunodeficiency Virus Type 1 Persistence Following Systemic Chemotherapy for Malignancy. J. Infect. Dis. 2017, 216, 254–262. [Google Scholar] [CrossRef] [Green Version]
- Buzon, M.J.; Sun, H.; Li, C.; Shaw, A.; Seiss, K.; Ouyang, Z.; Martin-Gayo, E.; Leng, J.; Henrich, T.J.; Li, J.Z.; et al. HIV-1 persistence in CD4+ T cells with stem cell-like properties. Nat. Med. 2014, 20, 139–142. [Google Scholar] [CrossRef]
- Schmueck-Henneresse, M.; Sharaf, R.; Vogt, K.; Weist, B.J.; Landwehr-Kenzel, S.; Fuehrer, H.; Jurisch, A.; Babel, N.; Rooney, C.M.; Reinke, P.; et al. Peripheral blood-derived virus-specific memory stem T cells mature to functional effector memory subsets with self-renewal potency. J. Immunol. 2015, 194, 5559–5567. [Google Scholar] [CrossRef] [Green Version]
- Barnes, P.D.; Grundy, J.E. Down-regulation of the class I HLA heterodimer and beta 2-microglobulin on the surface of cells infected with cytomegalovirus. J. Gen. Virol. 1992, 73, 2395–2403. [Google Scholar] [CrossRef]
- Ulbrecht, M.; Martinozzi, S.; Grzeschik, M.; Hengel, H.; Ellwart, J.W.; Pla, M.; Weiss, E.H. Cutting edge: The human cytomegalovirus UL40 gene product contains a ligand for HLA-E and prevents NK cell-mediated lysis. J. Immunol. 2000, 164, 5019–5022. [Google Scholar] [CrossRef] [Green Version]
- Lin, Y.L.; Chang, P.C.; Wang, Y.; Li, M. Identification of novel viral interleukin-10 isoforms of human cytomegalovirus AD169. Virus Res. 2008, 131, 213–223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- McSharry, B.P.; Avdic, S.; Slobedman, B. Human cytomegalovirus encoded homologs of cytokines, chemokines and their receptors: Roles in immunomodulation. Viruses 2012, 4, 2448–2470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hatano, H.; Jain, V.; Hunt, P.W.; Lee, T.H.; Sinclair, E.; Do, T.D.; Hoh, R.; Martin, J.N.; McCune, J.M.; Hecht, F.; et al. Cell-based measures of viral persistence are associated with immune activation and programmed cell death protein 1 (PD-1)-expressing CD4+ T cells. J. Infect. Dis. 2013, 208, 50–56. [Google Scholar] [CrossRef] [PubMed]
- Cooper, A.; Garcia, M.; Petrovas, C.; Yamamoto, T.; Koup, R.A.; Nabel, G.J. HIV-1 causes CD4 cell death through DNA-dependent protein kinase during viral integration. Nature 2013, 498, 376–379. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jakobsen, M.R.; Bak, R.O.; Andersen, A.; Berg, R.K.; Jensen, S.B.; Tengchuan, J.; Laustsen, A.; Hansen, K.; Ostergaard, L.; Fitzgerald, K.A.; et al. IFI16 senses DNA forms of the lentiviral replication cycle and controls HIV-1 replication. Proc. Natl. Acad. Sci. USA 2013, 110, E4571–E4580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kristoff, J.; Palma, M.L.; Garcia-Bates, T.M.; Shen, C.; Sluis-Cremer, N.; Gupta, P.; Rinaldo, C.R.; Mailliard, R.B. Type 1-programmed dendritic cells drive antigen-specific latency reversal and immune elimination of persistent HIV-1. EBiomedicine 2019, 43, 295–306. [Google Scholar] [CrossRef] [Green Version]
- Akiyama, Y.; Oshita, C.; Kume, A.; Iizuka, A.; Miyata, H.; Komiyama, M.; Ashizawa, T.; Yagoto, M.; Abe, Y.; Mitsuya, K.; et al. alpha-type-1 polarized dendritic cell-based vaccination in recurrent high-grade glioma: A phase I clinical trial. BMC Cancer 2012, 12, 623. [Google Scholar] [CrossRef] [Green Version]
- Fracol, M.; Xu, S.; Mick, R.; Fitzpatrick, E.; Nisenbaum, H.; Roses, R.; Fisher, C.; Tchou, J.; Fox, K.; Zhang, P.; et al. Response to HER-2 pulsed DC1 vaccines is predicted by both HER-2 and estrogen receptor expression in DCIS. Ann. Surg. Oncol. 2013, 20, 3233–3239. [Google Scholar] [CrossRef]
- Radomski, M.; Zeh, H.J.; Edington, H.D.; Pingpank, J.F.; Butterfield, L.H.; Whiteside, T.L.; Wieckowski, E.; Bartlett, D.L.; Kalinski, P. Prolonged intralymphatic delivery of dendritic cells through implantable lymphatic ports in patients with advanced cancer. J. Immunother. Cancer 2016, 4, 24. [Google Scholar] [CrossRef] [Green Version]
- Schuler, P.J.; Harasymczuk, M.; Visus, C.; Deleo, A.; Trivedi, S.; Lei, Y.; Argiris, A.; Gooding, W.; Butterfield, L.H.; Whiteside, T.L.; et al. Phase I dendritic cell p53 peptide vaccine for head and neck cancer. Clin. Cancer Res. 2014, 20, 2433–2444. [Google Scholar] [CrossRef] [Green Version]
- Feau, S.; Arens, R.; Togher, S.; Schoenberger, S.P. Autocrine IL-2 is required for secondary population expansion of CD8+ memory T cells. Nat. Immunol. 2011, 12, 908–913. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Feau, S.; Garcia, Z.; Arens, R.; Yagita, H.; Borst, J.; Schoenberger, S.P. The CD4+ T-cell help signal is transmitted from APC to CD8+ T-cells via CD27–CD70 interactions. Nat. Commun. 2012, 3, 948. [Google Scholar] [CrossRef] [PubMed]
- Estes, J.D.; Kityo, C.; Ssali, F.; Swainson, L.; Makamdop, K.N.; Del Prete, G.Q.; Deeks, S.G.; Luciw, P.A.; Chipman, J.G.; Beilman, G.J.; et al. Defining total-body AIDS-virus burden with implications for curative strategies. Nat. Med. 2017, 23, 1271–1276. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Santangelo, P.J.; Rogers, K.A.; Zurla, C.; Blanchard, E.L.; Gumber, S.; Strait, K.; Connor-Stroud, F.; Schuster, D.M.; Amancha, P.K.; Hong, J.J.; et al. Whole-body immunoPET reveals active SIV dynamics in viremic and antiretroviral therapy-treated macaques. Nat. Methods 2015, 12, 427–432. [Google Scholar] [CrossRef] [Green Version]
- Stieh, D.J.; Maric, D.; Kelley, Z.L.; Anderson, M.R.; Hattaway, H.Z.; Beilfuss, B.A.; Rothwangl, K.B.; Veazey, R.S.; Hope, T.J. Vaginal challenge with an SIV-based dual reporter system reveals that infection can occur throughout the upper and lower female reproductive tract. PLoS Pathog. 2014, 10, e1004440. [Google Scholar] [CrossRef]
- Zitvogel, L.; Regnault, A.; Lozier, A.; Wolfers, J.; Flament, C.; Tenza, D.; Ricciardi-Castagnoli, P.; Raposo, G.; Amigorena, S. Eradication of established murine tumors using a novel cell-free vaccine: Dendritic cell-derived exosomes. Nat. Med. 1998, 4, 594–600. [Google Scholar] [CrossRef]
Desired Trait | PGE2 DC | Type 1-polarized DC | References |
---|---|---|---|
Antigen Presentation | ++++ | ++++++ | [52] |
Maturation status (high CD83 / CD86) | ++++++ | ++++++ | [62,64] |
Lymph node homing (CCR7) | ++++++ | ++++ | [60,61,62,63,64] |
IL-12p70, IL-15 production | Deficient | ++++++++ | [62,64,72] |
T cell trafficking / chemokine expression | CCL22 (TREG) | CCL19 (TN, TCM) | [63,69,73,74] |
CCL3-5, CXCL9-11 (TEM) | |||
Antigen transfer ability | Deficient | ++++++ | [64] |
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kristoff, J.; Rinaldo, C.R.; Mailliard, R.B. Role of Dendritic Cells in Exposing Latent HIV-1 for the Kill. Viruses 2020, 12, 37. https://doi.org/10.3390/v12010037
Kristoff J, Rinaldo CR, Mailliard RB. Role of Dendritic Cells in Exposing Latent HIV-1 for the Kill. Viruses. 2020; 12(1):37. https://doi.org/10.3390/v12010037
Chicago/Turabian StyleKristoff, Jan, Charles R. Rinaldo, and Robbie B. Mailliard. 2020. "Role of Dendritic Cells in Exposing Latent HIV-1 for the Kill" Viruses 12, no. 1: 37. https://doi.org/10.3390/v12010037
APA StyleKristoff, J., Rinaldo, C. R., & Mailliard, R. B. (2020). Role of Dendritic Cells in Exposing Latent HIV-1 for the Kill. Viruses, 12(1), 37. https://doi.org/10.3390/v12010037