Supplementary Materials for

Similar documents
Supporting Information

Establishment and Targeting of the Viral Reservoir in Rhesus Monkeys

Decay characteristics of HIV-1- infected compartments during combination therapy

Supplementary Materials for

Combined IL-21 and IFNα treatment limits inflammation and delay viral rebound in ART-treated, SIV-infected macaques

Inves)gación básica y curación del VIH- 1

Approaching a Cure Daniel R. Kuritzkes, MD

Summary Report for HIV Random Clinical Trial Conducted in

Treatment with IL-7 Prevents the Decline of Circulating CD4 + T Cells during the Acute Phase of SIV Infection in Rhesus Macaques

Table S1. Viral load and CD4 count of HIV-infected patient population

Supporting Information

Simple Mathematical Models Do Not Accurately Predict Early SIV Dynamics

Supplementary Materials for

Induction of Innate Immune Responses in HVTN 071: a Trial using the MRKAd5 gag/pol/nef Vaccine from the Step Study

Supplementary Table 1 Clinicopathological characteristics of 35 patients with CRCs

<10. IL-1β IL-6 TNF + _ TGF-β + IL-23

Nature Medicine: doi: /nm.2109

Supplementary Materials for

Supplement for: CD4 cell dynamics in untreated HIV-1 infection: overall rates, and effects of age, viral load, gender and calendar time.

Supplementary Figure 1. ALVAC-protein vaccines and macaque immunization. (A) Maximum likelihood

Defining kinetic properties of HIV-specific CD8 + T-cell responses in acute infection

The potential role of PD-1/PD-L1 blockade in HIV Remission and Cure Strategies

Therapeutic Immunization with Autologous DC Pulsed with Autologous Inactivated HIV-1 Infected Apoptotic Cells

Therapeutic DNA Vaccine Induces Broad T Cell Responses in the Gut and Sustained Protection from Viral Rebound and AIDS in SIV-Infected Rhesus Macaques

IgG3 regulates tissue-like memory B cells in HIV-infected individuals

Supplementary Figure 1. Efficiency of Mll4 deletion and its effect on T cell populations in the periphery. Nature Immunology: doi: /ni.

Insight into treatment of HIV infection from viral dynamics models

SUPPLEMENTARY INFORMATION

HD1 (FLU) HD2 (EBV) HD2 (FLU)

Chronic HIV-1 Infection Frequently Fails to Protect against Superinfection

Defective STAT1 activation associated with impaired IFN-g production in NK and T lymphocytes from metastatic melanoma patients treated with IL-2

JAK3 inhibitor administration in vivo in chronically SIV Infected Rhesus Macaques

Clinical Development of ABX464, drug candidate for HIV Functional Cure. Chief Medical Officer ABIVAX

Interleukin-21 combined with ART reduces inflammation and viral reservoir in SIV-infected macaques

Mathematical-Statistical Modeling to Inform the Design of HIV Treatment Strategies and Clinical Trials

Figure S1. Alignment of predicted amino acid sequences of KIR3DH alleles identified in 8

B220 CD4 CD8. Figure 1. Confocal Image of Sensitized HLN. Representative image of a sensitized HLN

Ongoing HIV Replication During ART Reconsidered

Therapeutic Efficacy of a TLR7 Agonist for HBV Chronic Infection in Chimpanzees

Human Immunodeficiency Virus Type-1 Myeloid Derived Suppressor Cells Inhibit Cytomegalovirus Inflammation through Interleukin-27 and B7-H4

Analysis of left-censored multiplex immunoassay data: A unified approach

Viral Reservoirs and anti-latency interventions in nonhuman primate models of SIV/SHIV infection

5. Influence of multiplicity of infection (MOI) on synchronous baculovirus infections 5.1. Introduction

Immuno-modulatory Strategies for Reduction of HIV Reservoir Cells

Chapter 1: Exploring Data

Spleen. mlns. E Spleen 4.1. mlns. Spleen. mlns. Mock 17. Mock CD8 HIV-1 CD38 HLA-DR. Ki67. Spleen. Spleen. mlns. Cheng et al. Fig.

Low immune activation despite high levels of pathogenic HIV-1 results in long-term asymptomatic disease

CROI 2016 Review: Immunology and Vaccines

SCHOOL OF MATHEMATICS AND STATISTICS

ON ATTAINING MAXIMAL AND DURABLE SUPPRESSION OF THE VIRAL LOAD. Annah M. Jeffrey, Xiaohua Xia and Ian K. Craig

Supporting Information

Professor Jonathan Weber

VIRUS POPULATION DYNAMICS

Dynamics of lentiviral infection in vivo in the absence of adaptive immune responses

Rapid Seeding of the Viral Reservoir Prior to SIV Viremia in Rhesus Monkeys

Supplementary Figure 1. Enhanced detection of CTLA-4 on the surface of HIV-specific

Supplemental Information. CD4 + CD25 + Foxp3 + Regulatory T Cells Promote. Th17 Cells In Vitro and Enhance Host Resistance

Supplementary information. MARCH8 inhibits HIV-1 infection by reducing virion incorporation of envelope glycoproteins

Supplementary Figure 1. Gating strategy and quantification of integrated HIV DNA in sorted CD4 + T-cell subsets.

Models of HIV during antiretroviral treatment

Supplementary Figures. Supplementary Figure 1. Treatment schematic of SIV infection and ARV and PP therapies.

The human thymus is the central lymphoid organ that provides

Journal of Infectious Diseases Advance Access published July 11, Effect of Antiretroviral Therapy on HIV Reservoirs in Elite Controllers

Module R: Recording the HIV Reservoir

Understandable Statistics

ID Week 2016: HIV Update

Appendix III Individual-level analysis

Supplementary Figure 1

Major Depletion of Plasmacytoid Dendritic Cells in HIV-2 Infection, an Attenuated Form of HIV Disease

SUPPLEMENTARY INFORMATION

DEBATE ON HIV ENVELOPE AS A T CELL IMMUNOGEN HAS BEEN GAG-GED

Type and quantity of data needed for an early estimate of transmissibility when an infectious disease emerges

Supplemental Figure S1. Expression of Cirbp mrna in mouse tissues and NIH3T3 cells.

Human and mouse T cell regulation mediated by soluble CD52 interaction with Siglec-10. Esther Bandala-Sanchez, Yuxia Zhang, Simone Reinwald,

cure research HIV & AIDS

pplementary Figur Supplementary Figure 1. a.

well for 2 h at rt. Each dot represents an individual mouse and bar is the mean ±

Nature Genetics: doi: /ng Supplementary Figure 1. Rates of different mutation types in CRC.

Additional Presentation Demonstrates Potential Mechanisms for Unprecedented HIV Reservoir Depletion by SB-728-T

HIV cure: current status and implications for the future

Understanding HIV. Transmitted/Founder Viruses. Brandon Keele SAIC-Frederick National Cancer Institute

The Mechanisms for Within-Host Influenza Virus Control Affect Model-Based Assessment and Prediction of Antiviral Treatment

Development of 5 LTR DNA methylation of latent HIV-1 provirus in cell line models and in long-term-infected individuals

Supplementary information. Early development of broad neutralizing antibodies in HIV-1 infected infants

Supplementary Appendix

SUPPLEMENTARY INFORMATION. Divergent TLR7/9 signaling and type I interferon production distinguish

Supplementary Figure 1.

Supplementary Materials Extracting a Cellular Hierarchy from High-dimensional Cytometry Data with SPADE

Unit 1 Exploring and Understanding Data

NIH Public Access Author Manuscript J Acquir Immune Defic Syndr. Author manuscript; available in PMC 2013 September 01.

A VACCINE FOR HIV BIOE 301 LECTURE 10 MITALI BANERJEE HAART

MATERIALS AND METHODS. Neutralizing antibodies specific to mouse Dll1, Dll4, J1 and J2 were prepared as described. 1,2 All

Supplementary Materials

Imaging B Cell Follicles to Investigate HIV/SIV Persistence. Elizabeth Connick, M.D. University of Arizona May 8, 2017

Invited Review CROI 2018: Advances in Basic Science Understanding of HIV

An Introduction to Bayesian Statistics

Novel Approaches for the Assessment of the In Vivo Residual Virus Pool and Viral Eradication Strategies in SIV-infected Rhesus Macaques

Supplemental Table 1. Clinical and epidemiological characteristics of the

chapter 1 - fig. 2 Mechanism of transcriptional control by ppar agonists.

Transcription:

www.sciencetranslationalmedicine.org/cgi/content/full//49/eaao4/dc Supplementary Materials for TLR agonists induce transient viremia and reduce the viral reservoir in SIV-infected rhesus macaques on antiretroviral therapy So-Yon Lim, Christa E. Osuna, Peter T. Hraber, Joe Hesselgesser, Jeffrey M. Gerold, Tiffany L. Barnes, Srisowmya Sanisetty, Michael S. Seaman, Mark G. Lewis, Romas Geleziunas, Michael D. Miller, Tomas Cihlar, William A. Lee, Alison L. Hill, James B. Whitney* This PDF file includes: *Corresponding author. Email: jwhitne@bidmc.harvard.edu Published May 8, Sci. Transl. Med., eaao4 (8) DOI:.6/scitranslmed.aao4 Materials and Methods Table S. Parameters derived from viral dynamics models. Table S. Estimated number of latent cells reactivated during TLR agonist dosing period. Fig. S. Effective suppression of SIV replication after ART and study randomization. Fig. S. Study schema of TLR agonist dosing. Fig. S. Induction of IFN-α by TLR agonist dosing. Fig. S4. Cumulative gag and env phylogenies from GS-986 treated rhesus macaques. Fig. S. Env pseudovirus neutralization assay. Fig. S6. Changes in cell-associated viral DNA in mononuclear cells isolated from tissue compartments of rhesus macaques treated with a TLR agonist. Fig. S. The kinetics of SIV plasma RNA rebound after ART cessation. Fig. S8. Potent activation of CD6 + and CD6 CD6 NK and B cell subsets by TLR agonist exposure. Fig. S9. Magnitude of viral reactivation following repeated TLR agonist treatment. Fig. S. Induction of ISGs by TLR agonist dosing. Fig. S. Changes in cytokines and chemokines after TLR agonist administration.

Fig. S. Anamnestic SIV gag-specific CD4 + and CD8 + T cell responses after ART cessation in study. Fig. S. Coculture of CEMx4 cells with PBMCs after ART cessation. Fig. S4. In vivo CD8 + T lymphocyte depletion. Fig. S. Viral dynamics modeling of viral blips, rebound, and control. Fig. S6. Individual fits of viral dynamics model to viral load data.

Materials and Methods Viral dynamics modeling. We used viral dynamics models for three different purposes in the study: i) to characterize the kinetics of viral rebound after ART cessation among animals who rebounded, ii) to estimate the number of latent cells that would have needed to be reactivated to explain the blip dynamics seen during TLR-agonist + ART therapy, and iii) to estimate the latent reservoir reduction that would be needed to explain the lack of rebound in treated animals who did not rebound after ART cessation. (i) Modeling rebound kinetics after ART cessation: We fit each viral load time course to a standard model of viral dynamics (4) that describes the changes over time in levels of uninfected and infected target cells of the virus, along with free virus, using a system of ordinary differential equations (figure SA): T = λ βtv d T T I = a + βtv d I I V = ki cv where T is the concentration of uninfected target CD4 + T cells (cell/ul), I is the concentration of infected CD4 + T cells (cell/ml), V is the viral load (copies/ml), λ is the rate of production of susceptible uninfected cells (cells/ul/day), β is the viral infectivitiy ((copies/ml) - day - ), d I is the death rate of infected cells (day - ), d T is the death rate of death of uninfected cells (day - ), a is the rate at which latent cells reactivate to become productively infected(44-4), k is the viral burst size (virions/cell)(day - ) and c is the viral clearance rate (day - ). Because free virus is produced and cleared rapidly, levels are expected to equilibrate very rapidly with respect to infected cell levels, and we employed the common assumption that these two quantities are proportional. Consequently, the model reduces to: T = λ βtv d T T

V = a ( k c ) + βt (k c ) V d IV During ART, β =, and x and v reach a steady state at values T = λ/d T and V = (a/d I )(k/c), which we take as the initial conditions at the time of ART interruption. Note that this model assumes that some virus is present (due to stable release from reservoirs) immediately upon ART cessation and infection begins immediately. The reservoir exit rate a, and hence the initial value V is expected to be related to both the size and the activation rate of the functional viral reservoir. If the reservoir is so small, or so infrequently reactivating, that there is a delay between ART interruption and the start of the infection growth, this can be captured with very low a values. Separately, we used a stochastic model of functional reservoir activation to understand why some animals have not rebounded (see iii). The model was fit to longitudinal values of logarithmic plasma viral load (figure SB). Because direct measurements were not available for target cell levels (a subset of the total CD4 T cell population), the parameter λ and the combination (k/c) are not simultaneously identifiable. Accordingly, the ratio (k/c) was fixed to virus/cell based on a burst size of k = 4 (virions/cell)( day - ) and c= day - for SIV(-). In total, five parameters were fit (λ, β, a, d T, d I ). The inference procedure was formulated in a likelihood framework which treats data below the viral load detection threshold of copies/ml as censored. We assumed the observed viral load is log-normally distributed around the true viral load, which added an extra parameter for the measurement error variance σ. We maximized the likelihood over a log-transformed parameter space constrained by physiologically reasonable bounds: log(λ) [, ], log(β) [,], log(a) [ 8 ], log(d T ) [ 4, ], log ( d I d T ) [, ]. The differential equations were numerically integrated for each parameter value using ode4 and fits were obtained using the fminsearchbnd, both in MATLAB Ra (http://www.mathworks.com/matlabcentral/fileexchange/8-fminsearchbnd--fminsearchcon). For each animal, we started the fitting procedure using at least random initial condition values and chose the best resulting fit.

After fitting each model, we derived two composite parameters from model parameters the exponential growth rate of the virus immediately following rebound r, and the predicted set-point viral load, V SP (figure SC-F, Table S) In the realistic scenario where a is small and the residual viremia during ART is much less than viral loads off ART, these quantities are related to the model parameters by the following equations: r = λβ d T ( k c ) d I V SP = λ ( k d I c ) d T β In order to estimate how uncertainty in measurements of viral load data propagate to uncertainty in parameter estimates, we performed a perturbation analysis of our fitting procedure. Using the average error from our unperturbed fits of viral load in each animal on a logarithmic scale, we added noise to the viral load data above the detection limit by sampling from a normal distribution with matched variance and re-fit model parameters many times. All fits were visually inspected to ensure the minimization routine was not obviously caught in a local optimum. We performed statistical tests between treatment and control groups in order to ascertain statistically significant differences, which included parameter estimate uncertainties. All tests were performed using unpaired, two-sided, Welch's unequal variances t-tests(). The total sample variance for each group, s, was calculated as s = m + j p j /N, where m is the sample variance of the best estimated parameter values, and p j is the perturbation variance for each individual. Comparing parameters between pooled controls and pooled treated animals from both phases, we found that reactivation was lower in treated animals vs controls (mean log (a) -9.8 vs -.6 cell/ml/day, p=.), while viral growth rate was higher in treated animals vs controls (mean log (r). vs -. /day, corresponding to r of.8 vs./day, p=.). There was no significant difference in predicted set-point viral load or time to rebound. Comparing pooled controls to only those treated in Phase, the same trends held: in treated vs control animals, mean log (a) -. vs -.6 cell/ml/day (p=.) and mean log (r) -. vs -. /day (p=.). For Phase, pooling all controls, mean log (a) -9. vs -.6 cell/ml/day (p=.) and mean

log (r). vs -. /day (p=.). No significant differences were found in the other basic model parameters for either phase (p >.4), except in Phase, where the viral infectivity β, was significantly increased in treated animals (p =.). We repeated all statistical tests with Phase control animal - 8, as this animal fit least well with the model, and had very slow growth of viral load towards a set point without first reaching a peak. All the above trends were robust to removal of this outlier. We also repeated the model fitting assuming a washout period during which antiretroviral drugs were still decaying and the virus could not spread. Increasing this washout period to or days increased the estimated values of reactivation rate (a) but did not change the results of comparisons between groups. (ii) Modeling reservoir reactivation during TLR-agonist administration: To estimate the amount of latent cell reactivation needed to explain the observed blips, we used a similar model as above (figure SA), but assumed that β = during ART administration, and that the administration of the TLR-agonist caused the reactivation rate a to change to an amount a + a LRA. We assumed that a LRA was a constant, starting from a time t after TRL- agonist dosing began and continuing for a time τ. We allowed a LRA to vary between each animal and each dose. We used the same fitting procedure described in (i), except that an analytic formula for V(t) could be derived. We compared two scenarios for what happens when TLR-agonist action stops either reactivation ceases (a LRA ) but cells that are already reactivated continue to produce virions, or, virion production in reactivated cells also ceases. We found that the later scenario fit the blip data much better (figure SB). For this model, the equations for viral load are: ( k c ) ( a d I ) t < t V(t) = V(t ) + ( k c ) (a LRA ) ( ( ce dt(t t) d I e c(t t) )) t d I c d < t < τ I { V(t ) + V(τ)e ct t > τ

In all cases we fixed k = 4 (virions/cell) (day - ) and c= day - for SIV as above. To estimate d I for each animal, we fit the decay rate of viral load once ART was initiated to a simple exponential function (figure S6). We used a= - cell/ml/day which was the maximum value estimated above during rebound, though using alternate values of a that result in viral load below the detection limit before TLR-agonist dosing do not influence the results. Results depend to some extent on the values of d I, k, and c. If TLR-agonist-induced latent cells produce less virus over their lifespan than actively infected cells (e.g. due to incomplete activation), then the observed blips correspond to even higher numbers of actively infected cells. If they produce more virus (e.g. by avoiding immune detection), then blips could be explained with less reactivation. For each animal, the total reactivated cells over the entire treatment period was calculated as n_doses i= a ART,i τ i. In all TLR-agonist treated animals we found that between -4 cells/ml were likely activated which corresponds to.-% of SIV DNA+ PBMCs. (Supplemental Table ). Because other work has shown that for HIV only a few percent of DNA+ cells contain inducible or replication-competent virus(, ), it is plausible that a large fraction of the reservoir could have been reduced with these blip sizes. (iii) Modeling delay in viral rebound due to reservoir reduction. For animals who did not experience rebound, we were interested in how much the reservoir would need be have been reduced by the treatment to explain the long delay in rebound, assuming that this was the only mechanism for control. We used a previously developed stochastic model for HIV rebound dynamics(4) and adjusted the parameter values to represent the dynamics in SIVinfected macaques. Specifically, we reduced the total number of latently-infected cells before intervention by -fold, representing the approximate weight difference between macaques and humans, while keeping the infected frequency the same (~ IUPM). We increased the viral growth rate to r =.8 since viral loads grow more rapidly post-rebound in these animals (Table S, figure SD, (4)) compared to humans (who have r ~.4), and we adjusted the number of cells reactivating from the reservoir each day to ~ (compared to ~6 in humans) so that the average rebound time (to copies/ml) was around 8 days assuming three days for drug washout (Table S, figure SF, (4)). We kept the parameter determining the noise in viral burst size the

same and also assumed the death rate of latently infected cells was the same as in humans []. In keeping with our findings here and elsewhere (), we assume that there may be inter-individual variation in these parameters. In particular, we allowed the growth rate and reactivation rate to be log-normally distributed with log-means at the values given above, and log-standard deviations of. and. respectively. With these values, we ran the model to determine the distribution of expected rebound times (or probability of cure) as a function of the reduction in the reservoir conferred by treatment (figure SG). These results demonstrate that observable delays in rebound time are not expected unless there are large reductions in the size of the intact reservoir. Even robust reductions up to -fold in all animals may not be observable in rebound times unless cohorts are very large. Consequently, the observation of rebound in most TLR-agonist-treated animals does not rule out a modest but incomplete effect of the drug on the reservoir size. We next used a previously-developed Bayesian approach(6) to determine which reservoir reductions were most likely given the joint observations of a particular time off treatment ( years) and a negative co-culture assay (using 4 million cells)() (figure SH). We found that a,4- fold reduction in the reservoir is most likely given these findings (9% CI [, ]). Note that this model ignores the fact that improved immune function due to TLR-agonist treatment may also contribute to the control observed in two animals, and therefore the reservoir reductions estimates only apply in the case that this is the sole mechanism for rebound delay. Given that CD8+ T cell depletion did not lead to loss of control in these animals, we believe this is a reasonable hypothesis.

Table S. Parameters derived from viral dynamics models. Estimates and uncertainties for a, the rate of exit of actively infected cells from the latent reservoir; r, the exponential growth rate of virus immediately following rebound initiation; and V SP, the predicted eventual set-point viral load. Rebound time is the predicted time at which viral load crossed the rebound threshold of copies/ml. Best values are the best-estimated parameter fits using the measured data. STD values are the standard deviation of parameter values returned from the perturbation analysis. Parameter estimation and perturbation procedures are described in the Supplementary Methods. log(a) (c/ml) log(r) (day - ) log(vsp) (c/ml) Phase Dose ID Best STD Best STD Best STD Best Placebo -8 -.94 6. -.8.9 4..6 6. 6-8 -.8.8 -.. 4.98..8-9 -6..94.6... 8. 4-9 -8...9... 9. -9-4.. -...6. 6. Rebound time (days) GS-986 (.,., *.) mg/kg 4-9 -4.6. -.8...8. 6-8 -.9...4.9..6 66-8 -..96.....6 8-9 -.8.9.... 8.4-9 -.6.44.4..9. 9. Placebo 6-9 -..99.6.6 4.9.6 8.9 GS-986,.mg/kg - -9.... 4.6. 8. 8- -.6.9.. 4.. 8.4 88- -.8.9..4 4..4 9. 44- - - - - - - -

GS-96,. mg/kg GS-96,. mg/kg 9-9 -4.....9.. 9- -6..46...68. 9.6 4- -6..9..6 4.. 8. - - - - - - - - 4- -..8...8.. 4- -.6.4..6.6.44 9.4

Table S. Estimated number of latent cells reactivated during TLR agonist dosing period. A mathematical model of latent cell reactivation and virion production and clearance was fit to the viral load blips observed during the entire course of TLR-agonist treatment for both study Phases. Results of the model fit were used to estimate the total number of reactivated cells over all doses (Supplementary Methods). CD4 counts and SIV DNA levels in PBMC were measured immediately prior to TLR-agonist dosing. Phase Dose ID GS-986 (.,., *.) mg/kg Required SIV + cells reactivated in plasma to explain blip size [cell/ul] SIV DNA + cells / 6 CD4 + T cells CD4 count [cells/ul] 6-8. 86 668..% 66-8. 8.9.% 8-9.4 9 9.8.% -9. 8 4.9.% % of all SIV + CD4 + T cells reactivated to explain blip size [%] GS-986,.mg/kg 8-.6 8 44.6.6% 88-.6 8.8.% 44-. 6.8.9% GS-96,. mg/kg 9-9. 88 6..6% 9-. 8..% 4-.4 6 6.64.% GS-96,. mg/kg -. 46 6..4% 4-.8 4..% 4-. 9 8.49.%

A 9 8 Log SIV RNA copies/ml 6 4 B Log SIV RNA copies/ml SIV RNA copies AUC at ART initiation NS NS 8 6 4 SIV infection ART TLR 8 6 84 4 68 4 8 6 9 4 Time after SIV infection (Days) TLR Control TLR Control Days to SIV suppression 84 6 8 Time to suppression NS TLR Control Study TLR Control Figure S

Figure S. Effective suppression of SIV replication after ART and study randomization. Log plasma virus RNA was monitored in RMs from the day of SIVmac infection to the initiation of TLR agonist therapy (A). The initial viral burden and days to the complete viral suppression from the initiation of ART between groups of animals treated with TLR agonists or control were compared (B). Open and solid circles represent animals used in the Study -(TLR-treated, ; Control, ) and Study -(TLR-treated, ; Control, ) respectively. Plots show the median with interquartile range. The comparison of the values between groups was determined using a Mann-Whitney test.

A Study Placebo ART stop GS-986 escalating.-. mg/kg B Study Placebo EOW x months EOW x 9 GS-986. mg/kg EOW x months EOW x 9 GS-96. mg/kg EOW x months EOW x 9 ART stop GS-96. mg/kg EOW x months Figure S

Figure S. Study schema of TLR agonist dosing. In Study, 4 ART-suppressed monkeys received doses of GS-986: dose at. mg.kg, at. mg/kg, and then at. mg/kg. 6 control monkeys received formulation vehicle only (A). Doses were administered every other week (EOW). ART was discontinued weeks after the last TLR agonist dose. In Study, monkeys were distributed into 4 groups: in placebo (vehicle only), in GS-986 at. mg/kg, in GS-96 at. mg/kg, and in GS-96 at. mg/kg (B). doses were administered EOW followed by a -month period without dosing after which monkeys in the placebo, GS-986. mg/kg, and GS-96. mg/kg groups resumed dosing for 9 additional doses, again EOW. weeks after the last dose, ART was discontinued in all 4 groups.

A Log IFN-a (pg/ml) B 4 68 4 68 4 68 4 68 4 68 4 68 4 68 Control Time after TLR agonist dose (Hours) Dose - Dose -9 GS-986 (.mg/kg) Log IFN-α (pg/ml) GS-96 (.mg/kg) 4 4 4 4 4 4 4 4 4 4 GS-96 (.mg/kg) 4 4 4 4 4 4 4 4 4 Time after TLR agonist dose (Hours) Figure S

Figure S. Induction of IFN-α by TLR agonist dosing. Dose-dependent induction of plasma IFN-α in RMs treated with escalating doses of GS-986 (Study ) (A). Dose-dependent induction of IFN-α in RMs treated with GS-986 or GS-96 (Study ) (B). The dose concentrations of GS-986 or GS-96 are indicated. Shaded areas represent 4-hour post-dosing of TLR agonist.

gag nt Pre-TLR Blip Post-TLR env Pre-TLR Blip Post-TLR nt 6-8 66-8 -9 8-9 Pre-TLR Blip Post-TLR PB PL LN CR PB PL LN CR PB PL PB PL PB PL LN CR PB PL LN CR Figure S4A

gag env Pre-TLR Blip Post-TLR Pre-TLR Blip Post-TLR PL Hypermutated p<. Hypermutated p<. Hypermutated p<. PL Hypermutated p<. Hypermutated p<. Hypermutated p<. 6-8 PB LN PB LN CR CR PL PL 66-8 PB LN PB LN CR CR PL PL -9 PB LN PB LN CR CR PL PL 8-9 PB LN PB LN CR CR............ Evolutionary Distance to T/F Sequence, NT Substitutions per Site......... Evolutionary Distance to T/F Sequence, NT Substitutions per Site Figure S4B

Figure S4. Cumulative gag and env phylogenies from GS-986 treated rhesus macaques. (A) Cumulative phylogenies from four treated RMs: 6-8, 66-8, -9, and 8-9. For clarity, sequences that exactly matched the T/F sequence are excluded. Symbol shapes indicate the compartment from which the sequence was sampled: PL, cell-free plasma RNA; PB, cell-associated proviral DNA from peripheral blood mononuclear cells (PBMCs); CR, proviral colorectal DNA; and LN, proviral lymph node DNA. Sequences sampled pre-treatment appear in the left-hand column. The middle column additionally shows plasma and PBMC sequences sampled during viremic blips. The right column adds post-treatment sequences to the previous two. Symbol fill colors indicate when the sequence was sampled, whether solid (pre-gs-986 treatment), intermediate grey or red (viremic blip), or open (post-treatment). Hypermutated sequences (p<. by Hypermut v) are indicated by red symbols. Maximum-likelihood trees were obtained with phyml and GTR with gamma-four rate heterogeneity. Trees were rooted on the transmittedfounder sequence, taken as the consensus among pre-treatment sequences, then ladderized. For each RM, the scale is kept the same. (B) Cumulative gag and env distributions of genetic distances from the founder sequences in TLR agonist (GS-986) treated RMs. Sequences that exactly matched the SIV founder sequence were excluded, for clarity. This representation follows Figure S4A in layout and symbol representation. Rather than phylogenies, sequences are stratified by anatomical tissue. Sequences from pre-treatment (left column), then with added sequences from blip viremia (middle column), then post-treatment (right column), are included cumulatively. Symbol fill indicates sample timing. Superimposed lines indicate the quartiles ( percentile, median, and percentile) of each distribution. The x-axis uses a nonlinear (square-root transformed) scale to compare small distances.

A B RLU 4 6-8 66-8 8-9 -9 Cutoff RLU RLU 4 GS-986 Control Cutoff RLU 4 6 8 Dilution 4 6 8 Dilution Figure S

Figure S. Env pseudovirus neutralization assay. Panels of Env-pseudoviruses were produced to characterize the spectrum of their infectivity using a TZM-bl assay. SIV env sequences that were chosen to be cloned are isolates from SIV RNA blips (n=4) (A) or hypermutated viral DNA (n=) (B). Infectivity is expressed as relative light units (RLU).

Log SIV DNA copies/ 6 CD4+ memory cells PBMC P=.6 P=.4 P=.8 4 LNMC 4 P=. P=.4 P=. GMMC 4 P=. P=. P=. Study Study Pre TLR Post TLR GS-986 Control GS-986 GS-96 (.mg/kg) GS-96 (.mg/kg) Control P Pr e TLR Po st TLR P Pr e TLR Po st TLR P Pr e TLR Po st TLR Study Study Control Figure S6

Figure S6. Changes in cell-associated viral DNA in mononuclear cells isolated from tissue compartments of rhesus macaques treated with a TLR agonist. Proviral DNA (copies/ 6 CD4+ memory cells) was assessed at pre- and post-tlr agonist treatment in tissues. Each panel represents proviral DNA measured in peripheral blood mononuclear cells (PBMC), lymph node mononuclear cells (LNMC), and gut mucosa mononuclear cells (GMMC) in rhesus macaques. Changes in SIV DNA between pre- and post-treatment was compared using a Wilcoxon matched paired test.

A B Log SIV RNA copies/ml Log SIV RNA copies/ml 8 6 4 8 6 4 GS-986 Wk - Wk -8 AUC Control 8 6 4 GS-986 Control GS-986 Control Log AUC 4 8 4 49 6 Days post ART stop 9 8 6 4 GS-986 Control C Log SIV RNA copies/ml 8 6 4 GS-986 Wk - Wk -8 AUC P=. Control 8 6 4 GS-986 P=.4 Control Log AUC 9 8 6 4 GS-986 P=. Control Study (n=6) Controls (n=9) Figure S

Figure S. The kinetics of SIV plasma RNA rebound after ART cessation. SIV rebound kinetics following ART cessation was assessed (A). Median value of log SIV RNA copies/ml for each RM in both GS-986 treated (, n=4) and control ( shown relative to the day of ART stop (Day ). One animal from control group with low viremia was indicated as an outlier (, n=6) groups with nine additional control RMs ( ) is ). Log plasma virus RNA was assessed in SIV-infected RMs between days to 6 following ART stop (B). Compasiron of TLR and control group from study and with additional nine control RMs (C) is shown. The plasma SIV RNA was assessed between days 4- (Week -) and 49-6 (Week -8) following ART cessation, representing peak and set-point viral load, respectively. Area under the curve calculations for the plasma SIV RNA burden were assessed though days 6 post ATI. The results are expressed as the median with interquartile range. The comparison between groups was determined using a Mann- Whitney test. 4 GS-986 Control

A Dose - Dose -9 6 4 CD6+ CD6- CD6+ 6 4 CD6+ CD6- CD6- CD6- CD6+ CD6- CD6-6 6 4 4 % CD69 + (difference) 6 4 6 4 6 4 Dose: 4 6 8 9 4 6 8 9 4 6 8 9 Vehicle GS-96. mg/kg GS-986 GS-96. mg/kg Dose: Figure S8A

B Dose - Dose -9 Naive Memory Naive Memory CD8 GMF (difference) Dose: 4 6 8 9 Vehicle GS-96. mg/kg 4 6 8 9 GS-986 GS-96. mg/kg Dose: Figure S8B

Figure S8. Potent activation of CD6 + and CD6 CD6 NK and B cell subsets by TLR agonist exposure. (A) Activation of CD6 +, CD6 - CD6 +, and CD6 - CD6 - NK cells in the peripheral blood of vehicle ( ), GS-986,. mg/kg ( ), GS-96,. mg/kg ( ), and GS-96,. mg/kg ( ) treated monkeys was monitored by flow cytometric detection of CD69. CD69 expression within each subset was measured at time of dose and at 4,, and (doses - only) hours post-dose (time points indicated by x-axis ticks) for doses,,, and (left) and doses -9 (right). Shown are the differences in percent CD69 + cells compared to day of dose. (B) Modulation of B cells by TLR agonists. Activation of naïve (CD - ) and memory (CD + ) B cells in the peripheral blood of vehicle ( ), GS-986,. mg/kg ( ), GS-96,. mg/kg ( ), and GS-96,. mg/kg ( ) treated monkeys was monitored by flow cytometric detection of CD8. CD8 geometric mean fluorescence (GMF) expression within each subset was measured at time of dose and at 4,, and (doses - only) hours post dose (time points indicated by x-axis ticks) for doses,,, and (left) and doses -9 (right). Shown are the differences in normalized CD8 GMF compared to day of dose.

A AUC 4....... 4 6 TLR agonist dose (mg/kg) B AUC 4 8 Control GS-986 (.mg/kg) GS96 (.mg/kg) GS-96 (.mg/kg) 9 9 9 9 TLR agonist dose Figure S9

Figure S9. Magnitude of viral reactivation following repeated TLR agonist treatment. The AUC of SIV plasma RNA copies was used as a measure of the total amount of SIV reactivation occurring in RMs during - h post each TLR agonist dose. A. RMs were treated with escalating doses of GS- 986. B. Groups of RMs treated with vehicle (control),. mg/kg GS-986,. mg/kg or. mg/kg of GS-96 are shown. Line and error bars represent the mean and standard deviation (SD).

ISGs Group ISGs Group ISG Mx OAS ISG Mx OAS Group ISG Fold Changes Whitney J. Unpublished Data Baseline 4 4 4 4 4 4 4 4 4 Vehicle 4 6 TLR agonist Supplementary figure 6A.. The peak induction of each ISG in monkeys in either group or 4 was observed within 4h, and usually returned to baseline within h. Figure S. GS46986 was the most potent TLR agonist to induce highest mrna expression of ISGs.

Figure S. Induction of ISGs by TLR agonist dosing. Heatmap depiction of gene expression patterns of ISG-, Mx- and OAS- in RMs from vehicle (n=), GS-986,.mg/kg (n=), GS-96,.mg/kg (n=) and GS-96,.mg/kg (n=) groups. Values reflect fold changes of mrna expression of three ISGs at 4,, and hours after each TLR agonist dose compared to that of baseline are shown by group and number of TLR doses.

A IL-RA IL-6 TGF-β TNF-α GM-CSF IL-8 MCP- RANTES scd4l I-TAC Log Fold Changes - - 4 6 4 6 4 6 4 6 4 6 4 6 TLR agonist dose 4 6 4 6 4 6 4 6 B 4 IL-RA_ -Log P value TNFα_6 TNFα_ I-TAC_ I-TAC_ I-TAC_6 IL-RA_ IL-RA_6 IL-RA_4 IL-RA_ IL-RA_ IL-RA_ - - - Log fold change Figure SAB

C Log Fold Changes 4 - - 4 - - 4 - - 4 - - IL-RA IL-8 IL-6 RANTES Control GS-986 GS-96 (. mg.kg) GS-96 (. mg.kg) D -Log P value 4 Control GS-986 GS-96 (. mg/kg) GS-96 (. mg/kg) IL-RA_ I-TAC_4 I-TAC_6 I-TAC_ IL-RA_ IL-RA_ I-TAC_ IL-RA_4 I-TAC_ - - - Log fold change IL-RA_ 4 - - I-TAC 4 - - scd4l 9 9 8 4 6 8 9 9 8 4 6 8 4 6 4 6 TLR agonist dose TLR dose Figure SCD

Figure S. Changes in cytokines and chemokines after TLR agonist administration. Changes in the level of various inflammatory cytokines and chemokines were measured in plasma between pre- and 4 hours after each TLR agonist dose. Values reflecting fold changes of the amounts of inflammatory cytokines and chemokines detected in plasma isolated from four monkeys treated with escalating doses of TLR agonist (GS-986) (A) and the association of TLR doses with cytokine/chemokine induction was assessed in groups of monkeys treated with escalating doses of GS-986 or vehicle (control) (B) Fold changes in the amounts of cytokines and chemokines indicated in groups of monkeys treated with vehicle (control), GS-986 (. mg/kg) and GS-96 (. mg/kg,. mg/kg) compared to baseline are shown (C) Association of TLR agonist doses and the amount of cytokine and chemokine induced is shown (D) Values are expressed as mean ± standard deviation (SD). Validated ELISA assays were used to determine concentration of cytokines and chemokines including IL-RA, IL-6, TNF-α, GM-CSF, TGF-β, IL-8, I-TAC, MCP-, RANTES, and scd4l as described in Methods. Association of TLR dose responses with cytokine/chemokine induction was assessed in groups of monkeys treated with escalating doses of GS-986 (C, Study ), or. mg/kg of GS-986,. mg/kg or. mg/kg of GS96 (D, Study ). Volcano plots show up/down regulation of cytokine and chemokine levels as response to TLR agonist dose. The relative ratio of cytokine and chemokines levels after TLR dose was compared to that of the base line. Values that showed statistically significant changes from baseline are shown in red.

. CD4+ T Cells. CD8+ T Cells Control-Viremic GS-986-Viremic GS-986-Aviremic % IFNg+... % IFNg+.. GS-96 (. mg/ml)-viremic GS-96 (. mg/ml)-viremic GS-96 (. mg/ml)-aviremic. Time after ART stop (Days). Time after ART stop (Days) Figure S

Figure S. Anamnestic SIV gag-specific CD4 + and CD8 + T cell responses after ART cessation in study. Shown are the frequencies of IFN-γproducing SIV Gag-specific CD4+ and CD8+ T cells, measured by intracellular cytokine staining, on the day of ART stop (day ) and on days and after stop. The background-subtracted percent of IFN-γ-positive CD4+ T (left panel) and CD8+ T (right panel) are shown for monkeys that became viremic (closed symbols) or remained aviremic (open symbols) in each treatment group.

A B Log SIV RNA copies/ml Log SIV RNA copies/ml 8 6 4 Control No stimulation ConA 9 GS-986 (.mg/kg) No stimulation ConA GS-96 (. mg/kg) No stimulation ConA 9 4 Days post co-culture Control GS-986 GS-96 (.mg/kg) Figure S

Figure S. Coculture of CEMx4 cells with PBMCs after ART cessation. PBMC were isolated from two control RMs (6-9 and -) and two aviremic RMs (- and 44-) on days 8 post ATI and used in viral outgrowth assay (A) and in vitro co-culture assay with susceptible CEMx4 cells following CD8+ T cell depletion (B).

Days post CD8 depletion 4 8 88-9- CD8 44- - CD Figure S4

Figure S4. In vivo CD8 + T lymphocyte depletion. CD8+ lymphocytes were depleted in vivo using monoclonal antibody MT8R in two viremic animals (88- and 9-) and the two aviremic RMs (- and 44-). The presence of CD8+ lymphocytes in the peripheral blood was monitored by flow cytometry. The plots of CD8 and CD fluorescent antibody staining up to day post depletion are shown.

Figure S. Viral dynamics modeling of viral blips, rebound, and control. Schematic of the viral dynamics model used to describe i) viral decay after ART initiation, ii) viral production from latently infected cells during TLR-agonist administration, and iii) viral rebound following ART cessation (A). We assume ART inhibits viral infectivity, and TLR-agonist transiently stimulates reactivation of latent cells. Summary of the differences in rebound kinetics between treated and control animals (B). Each curve is generated using the geometric mean of the parameter values from each group. Values of viral dynamics parameters during rebound estimated from model fits (C - F). Control and treatment animals from both study phases were pooled for comparisons between groups. Statistical tests took into account the uncertainty in parameter estimates from each animal. The distribution of rebound times was predicted as a function of the reduction in the reservoir size, using a separate stochastic model of viral reactivation and rebound (G). Survival curves show the predicted percentage of animals that have not yet rebounded as a function of time after ART cessation. The median posterior probability (and 9% credible interval) for the reservoir reduction caused by treatment, as a function of the current time off ART without rebound (H). We assume a baseline reservoir size of ~ IUPM. At the time of ART interruption, a negative viral outgrowth assay suggests that the reservoir is decreased by.8 [.-.9] log. After 6 months without rebound, the updated estimate for the reservoir reduction is.6 [.-4.] logs, and after years the estimate is. [.-4.] logs (or,4 [-,] fold decrease). Details of all the modeling are described in the Supplementary Methods.

Viral load (log RNA copies/ml) // 4 4 6 6 6-8 // 4 4 6 6 6-8 // 4 4 6 6 66-8 // 4 4 6 6 8-9 // 4 4 6 6-9 Study -8 // 4 4 6 6-9 // 4 4 6 6 4-9 // 4 4 6 6-9 // 4 4 6 6 4-9 // 4 4 6 6 data model fit control treated ART stop Time after ART initiation (days) Study ART stop 6-9 // ` 4 4 6 6 8 8 9 9 - // 4 4 6 6 8 8 9 9 8- // ` 4 4 6 6 8 8 9 9 88- // 4 4 6 6 8 8 9 9 44- // 4 4 6 6 8 8 9 9 9-9 // 4 4 6 6 8 8 9 9 9- // 4 4 6 6 8 8 9 9 4- // 4 4 6 6 8 8 9 9 - // 4 4 6 6 8 8 9 9 4- // 4 4 6 6 8 8 9 9 4- // 4 4 6 6 8 8 9 9 Time after ART initiation (days) Figure S6

Figure S6. Individual fits of viral dynamics model to viral load data. Time course of observed viral dynamics (black dots) and model fits (solid lines) for control (red) and treated (blue) animals. Viral load is shown from the time ART is initiated until the end of the study period. Dots are shown at each time point at which viral load was measured. Model fits were generated using the maximum likelihood estimate for each parameter value. Separate models were used to fit the time from ART initiation to undetectable viremia, viral blips, and viral rebound (see Supplementary Methods for details).