Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Morgane Rolland is active.

Publication


Featured researches published by Morgane Rolland.


Nature | 2012

Increased HIV-1 vaccine efficacy against viruses with genetic signatures in Env V2

Morgane Rolland; Paul T. Edlefsen; Brendan B. Larsen; Sodsai Tovanabutra; Eric Sanders-Buell; Tomer Hertz; Allan C. deCamp; Chris Carrico; Sergey Menis; Craig A. Magaret; Hasan Ahmed; Michal Juraska; Lennie Chen; Philip Konopa; Snehal Nariya; Julia N. Stoddard; Kim Wong; Haishuang Zhao; Wenjie Deng; Brandon Maust; Meera Bose; Shana Howell; A Bates; Michelle Lazzaro; Annemarie O'Sullivan; Esther Lei; Andrea Bradfield; Grace Ibitamuno; Vatcharain Assawadarachai; Robert J. O'Connell

The RV144 trial demonstrated 31% vaccine efficacy at preventing human immunodeficiency virus (HIV)-1 infection. Antibodies against the HIV-1 envelope variable loops 1 and 2 (Env V1 and V2) correlated inversely with infection risk. We proposed that vaccine-induced immune responses against V1/V2 would have a selective effect against, or sieve, HIV-1 breakthrough viruses. A total of 936 HIV-1 genome sequences from 44 vaccine and 66 placebo recipients were examined. We show that vaccine-induced immune responses were associated with two signatures in V2 at amino acid positions 169 and 181. Vaccine efficacy against viruses matching the vaccine at position 169 was 48% (confidence interval 18% to 66%; P = 0.0036), whereas vaccine efficacy against viruses mismatching the vaccine at position 181 was 78% (confidence interval 35% to 93%; P = 0.0028). Residue 169 is in a cationic glycosylated region recognized by broadly neutralizing and RV144-derived antibodies. The predicted distance between the two signature sites (21 ± 7 Å) and their match/mismatch dichotomy indicate that multiple factors may be involved in the protection observed in RV144. Genetic signatures of RV144 vaccination in V2 complement the finding of an association between high V1/V2-binding antibodies and reduced risk of HIV-1 acquisition, and provide evidence that vaccine-induced V2 responses plausibly had a role in the partial protection conferred by the RV144 regimen.


Nature Medicine | 2011

Genetic impact of vaccination on breakthrough HIV-1 sequences from the STEP trial

Morgane Rolland; Sodsai Tovanabutra; Allan C. deCamp; Nicole Frahm; Peter B. Gilbert; Eric Sanders-Buell; Laura Heath; Craig A. Magaret; Meera Bose; Andrea Bradfield; Annemarie O'Sullivan; Jacqueline Crossler; Teresa Jones; Marty Nau; Kim Wong; Hong Zhao; Dana N. Raugi; Stephanie Sorensen; Julia N. Stoddard; Brandon Maust; Wenjie Deng; John Hural; Sheri A. Dubey; Nelson L. Michael; John W. Shiver; Lawrence Corey; Fusheng Li; Steve Self; Jerome H. Kim; Susan Buchbinder

We analyzed HIV-1 genome sequences from 68 newly infected volunteers in the STEP HIV-1 vaccine trial. To determine whether the vaccine exerted selective T cell pressure on breakthrough viruses, we identified potential T cell epitopes in the founder sequences and compared them to epitopes in the vaccine. We found greater distances to the vaccine sequence for sequences from vaccine recipients than from placebo recipients. The most significant signature site distinguishing vaccine from placebo recipients was Gag amino acid 84, a site encompassed by several epitopes contained in the vaccine and restricted by human leukocyte antigen (HLA) alleles common in the study cohort. Moreover, the extended divergence was confined to the vaccine components of the virus (HIV-1 Gag, Pol and Nef) and not found in other HIV-1 proteins. These results represent what is to our knowledge the first evidence of selective pressure from vaccine-induced T cell responses on HIV-1 infection in humans.


Science Translational Medicine | 2014

Polyfunctional Fc-Effector Profiles Mediated by IgG Subclass Selection Distinguish RV144 and VAX003 Vaccines

Amy W. Chung; Musie Ghebremichael; Hannah Robinson; Eric P. Brown; Ickwon Choi; Sophie Lane; Anne-Sophie Dugast; Matthew K. Schoen; Morgane Rolland; Todd J. Suscovich; Alison E. Mahan; Larry Liao; Hendrik Streeck; Charla Andrews; Supachai Rerks-Ngarm; Sorachai Nitayaphan; Mark S. de Souza; Jaranit Kaewkungwal; Punnee Pitisuttithum; Donald P. Francis; Nelson L. Michael; Jerome H. Kim; Chris Bailey-Kellogg; Margaret E. Ackerman; Galit Alter

RV144 vaccination induced polyfunctional antibody Fc-effector responses, whereas VAX003 vaccination increased inhibitory IgG4 antibodies. More Is Better for Protection Against HIV Recently, results from the first protective HIV phase 2B RV144 vaccine trial pointed to an unexpected signature of protection, not associated with the traditional mechanisms of vaccine-induced immunity, namely, neutralizing antibodies and killer T cell immunity. Instead, protection was associated with specific subpopulations of antibodies that were able to direct killing of HIV-infected cells. However, little is known about the properties of these killer antibodies or their biophysical features. In a new study, Chung et al. functionally profiled antibodies raised by the protective RV144 vaccine trial and its nonprotective predecessor, the VAX003 vaccine trial, both conducted in Thailand. RV144 vaccination uniquely induced antibodies capable of directing several different antiviral functions in a coordinated manner. In contrast, VAX003 vaccination predominantly induced single or uncoordinated antiviral responses. Functional coordination was regulated by the selection of antibody responses directed at vulnerable regions on the HIV envelope that were specifically tuned to enhanced functionality through the selection of a specific antibody subclass, IgG3, known to harbor strong antiviral activity. Collectively, these data suggest that vaccines able to induce broader antibody functional profiles, through the selection of more potent antibody subclasses, which target vulnerable regions of the virus, may represent a new means by which to achieve protection from HIV infection in the absence of neutralization. The human phase 2B RV144 ALVAC-HIV vCP1521/AIDSVAX B/E vaccine trial, held in Thailand, resulted in an estimated 31.2% efficacy against HIV infection. By contrast, vaccination with VAX003 (consisting of only AIDSVAX B/E) was not protective. Because protection within RV144 was observed in the absence of neutralizing antibody activity or cytotoxic T cell responses, we speculated that the specificity or qualitative differences in Fc-effector profiles of nonneutralizing antibodies may have accounted for the efficacy differences observed between the two trials. We show that the RV144 regimen elicited nonneutralizing antibodies with highly coordinated Fc-mediated effector responses through the selective induction of highly functional immunoglobulin G3 (IgG3). By contrast, VAX003 elicited monofunctional antibody responses influenced by IgG4 selection, which was promoted by repeated AIDSVAX B/E protein boosts. Moreover, only RV144 induced IgG1 and IgG3 antibodies targeting the crown of the HIV envelope V2 loop, albeit with limited coverage of breakthrough viral sequences. These data suggest that subclass selection differences associated with coordinated humoral functional responses targeting strain-specific protective V2 loop epitopes may underlie differences in vaccine efficacy observed between these two vaccine trials.


Journal of Virology | 2008

Central Role of Reverting Mutations in HLA Associations with Human Immunodeficiency Virus Set Point

Philippa C. Matthews; Andrew J. Prendergast; Alasdair Leslie; Hayley Crawford; Rebecca Payne; Christine Rousseau; Morgane Rolland; Isobella Honeyborne; Jonathan M. Carlson; Carl M. Kadie; Christian Brander; Karen Bishop; Nonkululeko Mlotshwa; James I. Mullins; Hoosen Coovadia; Thumbi Ndung'u; Bruce D. Walker; David Heckerman; Philip J. R. Goulder

ABSTRACT Much uncertainty still exists over what T-cell responses need to be induced by an effective human immunodeficiency virus (HIV) vaccine. Previous studies have hypothesized that the effective CD8+ T-cell responses are those driving the selection of escape mutations that reduce viral fitness and therefore revert posttransmission. In this study, we adopted a novel approach to define better the role of reverting escape mutations in immune control of HIV infection. This analysis of sequences from 710 study subjects with chronic C-clade HIV type 1 infection demonstrates the importance of mutations that impose a fitness cost in the control of viremia. Consistent with previous studies, the viral set points associated with each HLA-B allele are strongly correlated with the number of Gag-specific polymorphisms associated with the relevant HLA-B allele (r = −0.56, P = 0.0034). The viral set points associated with each HLA-C allele were also strongly correlated with the number of Pol-specific polymorphisms associated with the relevant HLA-C allele (r = −0.67, P = 0.0047). However, critically, both these correlations were dependent solely on the polymorphisms identified as reverting. Therefore, despite the inevitable evolution of viral escape, viremia can be controlled through the selection of mutations that are detrimental to viral fitness. The significance of these results is in highlighting the rationale for an HIV vaccine that can induce these broad responses.


PLOS ONE | 2008

Broad and Gag-Biased HIV-1 Epitope Repertoires Are Associated with Lower Viral Loads

Morgane Rolland; David Heckerman; Wenjie Deng; Christine Rousseau; Hoosen Mohamed Coovadia; Karen Bishop; Philip J. R. Goulder; Bruce D. Walker; Christian Brander; James I. Mullins

Background HLA class-I alleles differ in their ability to control HIV replication through cell-mediated immune responses. No consistent associations have been found between the breadth of Cytotoxic T Lymphocytes (CTL) responses and the control of HIV-1, and it is unknown whether the size or distribution of the viral proteome-wide epitope repertoire, i.e., the intrinsic ability to present fewer, more or specific viral epitopes, could affect clinical markers of disease progression. Methodology/Principal Findings We used an epitope prediction model to identify all epitope motifs in a set of 302 HIV-1 full-length proteomes according to each individuals HLA (Human Leukocyte Antigen) genotype. The epitope repertoire, i.e., the number of predicted epitopes per HIV-1 proteome, varied considerably between HLA alleles and thus among individual proteomes. In a subgroup of 270 chronically infected individuals, we found that lower viral loads and higher CD4 counts were associated with a larger predicted epitope repertoire. Additionally, in Gag and Rev only, more epitopes were restricted by alleles associated with low viral loads than by alleles associated with higher viral loads. Conclusions/Significance This comprehensive analysis puts forth the epitope repertoire as a mechanistic component of the multi-faceted HIV-specific CTL response. The favorable impact on markers of disease status of the propensity to present more HLA binding peptides and specific proteins gives impetus to vaccine design strategies that seek to elicit responses to a broad array of HIV-1 epitopes, and suggest a particular focus on Gag.


Journal of Virology | 2008

HLA Class I-Driven Evolution of Human Immunodeficiency Virus Type 1 Subtype C Proteome: Immune Escape and Viral Load

Christine Rousseau; Marcus Daniels; Jonathan M. Carlson; Carl M. Kadie; Hayley Crawford; Andrew J. Prendergast; Philippa C. Matthews; Rebecca Payne; Morgane Rolland; Dana N. Raugi; Brandon Maust; Gerald H. Learn; David C. Nickle; Hoosen Coovadia; Thumbi Ndung'u; Nicole Frahm; Christian Brander; Bruce D. Walker; Philip J. R. Goulder; Tanmoy Bhattacharya; David Heckerman; Bette Korber; James I. Mullins

ABSTRACT Human immunodeficiency virus type 1 (HIV-1) mutations that confer escape from cytotoxic T-lymphocyte (CTL) recognition can sometimes result in lower viral fitness. These mutations can then revert upon transmission to a new host in the absence of CTL-mediated immune selection pressure restricted by the HLA alleles of the prior host. To identify these potentially critical recognition points on the virus, we assessed HLA-driven viral evolution using three phylogenetic correction methods across full HIV-1 subtype C proteomes from a cohort of 261 South Africans and identified amino acids conferring either susceptibility or resistance to CTLs. A total of 558 CTL-susceptible and -resistant HLA-amino acid associations were identified and organized into 310 immunological sets (groups of individual associations related to a single HLA/epitope combination). Mutations away from seven susceptible residues, including four in Gag, were associated with lower plasma viral-RNA loads (q < 0.2 [where q is the expected false-discovery rate]) in individuals with the corresponding HLA alleles. The ratio of susceptible to resistant residues among those without the corresponding HLA alleles varied in the order Vpr > Gag > Rev > Pol > Nef > Vif > Tat > Env > Vpu (Fishers exact test; P ≤ 0.0009 for each comparison), suggesting the same ranking of fitness costs by genes associated with CTL escape. Significantly more HLA-B (χ2; P = 3.59 × 10−5) and HLA-C (χ2; P = 4.71 × 10−6) alleles were associated with amino acid changes than HLA-A, highlighting their importance in driving viral evolution. In conclusion, specific HIV-1 residues (enriched in Vpr, Gag, and Rev) and HLA alleles (particularly B and C) confer susceptibility to the CTL response and are likely to be important in the development of vaccines targeted to decrease the viral load.


PLOS Medicine | 2015

Geographic and Temporal Trends in the Molecular Epidemiology and Genetic Mechanisms of Transmitted HIV-1 Drug Resistance: An Individual-Patient- and Sequence-Level Meta-Analysis

Soo Yon Rhee; Jose L. Blanco; Michael R. Jordan; Jonathan Taylor; Philippe Lemey; Vici Varghese; Raph L. Hamers; Silvia Bertagnolio; Tobias F. Rinke de Wit; Avelin F. Aghokeng; Jan Albert; Radko Avi; Santiago Avila-Rios; Pascal Bessong; James Brooks; Charles A. Boucher; Zabrina L. Brumme; Michael P. Busch; Hermann Bussmann; Marie Laure Chaix; Bum Sik Chin; Toni T. D’Aquin; Cillian F. De Gascun; Anne Derache; Diane Descamps; Alaka Deshpande; Cyrille F. Djoko; Susan H. Eshleman; Hervé Fleury; Pierre Frange

Background Regional and subtype-specific mutational patterns of HIV-1 transmitted drug resistance (TDR) are essential for informing first-line antiretroviral (ARV) therapy guidelines and designing diagnostic assays for use in regions where standard genotypic resistance testing is not affordable. We sought to understand the molecular epidemiology of TDR and to identify the HIV-1 drug-resistance mutations responsible for TDR in different regions and virus subtypes. Methods and Findings We reviewed all GenBank submissions of HIV-1 reverse transcriptase sequences with or without protease and identified 287 studies published between March 1, 2000, and December 31, 2013, with more than 25 recently or chronically infected ARV-naïve individuals. These studies comprised 50,870 individuals from 111 countries. Each set of study sequences was analyzed for phylogenetic clustering and the presence of 93 surveillance drug-resistance mutations (SDRMs). The median overall TDR prevalence in sub-Saharan Africa (SSA), south/southeast Asia (SSEA), upper-income Asian countries, Latin America/Caribbean, Europe, and North America was 2.8%, 2.9%, 5.6%, 7.6%, 9.4%, and 11.5%, respectively. In SSA, there was a yearly 1.09-fold (95% CI: 1.05–1.14) increase in odds of TDR since national ARV scale-up attributable to an increase in non-nucleoside reverse transcriptase inhibitor (NNRTI) resistance. The odds of NNRTI-associated TDR also increased in Latin America/Caribbean (odds ratio [OR] = 1.16; 95% CI: 1.06–1.25), North America (OR = 1.19; 95% CI: 1.12–1.26), Europe (OR = 1.07; 95% CI: 1.01–1.13), and upper-income Asian countries (OR = 1.33; 95% CI: 1.12–1.55). In SSEA, there was no significant change in the odds of TDR since national ARV scale-up (OR = 0.97; 95% CI: 0.92–1.02). An analysis limited to sequences with mixtures at less than 0.5% of their nucleotide positions—a proxy for recent infection—yielded trends comparable to those obtained using the complete dataset. Four NNRTI SDRMs—K101E, K103N, Y181C, and G190A—accounted for >80% of NNRTI-associated TDR in all regions and subtypes. Sixteen nucleoside reverse transcriptase inhibitor (NRTI) SDRMs accounted for >69% of NRTI-associated TDR in all regions and subtypes. In SSA and SSEA, 89% of NNRTI SDRMs were associated with high-level resistance to nevirapine or efavirenz, whereas only 27% of NRTI SDRMs were associated with high-level resistance to zidovudine, lamivudine, tenofovir, or abacavir. Of 763 viruses with TDR in SSA and SSEA, 725 (95%) were genetically dissimilar; 38 (5%) formed 19 sequence pairs. Inherent limitations of this study are that some cohorts may not represent the broader regional population and that studies were heterogeneous with respect to duration of infection prior to sampling. Conclusions Most TDR strains in SSA and SSEA arose independently, suggesting that ARV regimens with a high genetic barrier to resistance combined with improved patient adherence may mitigate TDR increases by reducing the generation of new ARV-resistant strains. A small number of NNRTI-resistance mutations were responsible for most cases of high-level resistance, suggesting that inexpensive point-mutation assays to detect these mutations may be useful for pre-therapy screening in regions with high levels of TDR. In the context of a public health approach to ARV therapy, a reliable point-of-care genotypic resistance test could identify which patients should receive standard first-line therapy and which should receive a protease-inhibitor-containing regimen.


PLOS ONE | 2012

CTL Responses of High Functional Avidity and Broad Variant Cross-Reactivity Are Associated with HIV Control

Beatriz Mothe; Anuska Llano; Javier Ibarrondo; Jennifer Zamarreño; Mattia Schiaulini; Cristina Miranda; Marta Ruiz-Riol; Christoph T. Berger; M. José Herrero; Eduard Palou; Montse Plana; Morgane Rolland; Ashok Khatri; David Heckerman; Florencia Pereyra; Bruce D. Walker; David B. Weiner; Roger Paredes; Bonaventura Clotet; Barbara K. Felber; George N. Pavlakis; James I. Mullins; Christian Brander

Cytotoxic T lymphocyte (CTL) responses targeting specific HIV proteins, in particular Gag, have been associated with relative control of viral replication in vivo. However, Gag-specific CTL can also be detected in individuals who do not control the virus and it remains thus unclear how Gag-specific CTL may mediate the beneficial effects in some individuals but not in others. Here, we used a 10mer peptide set spanning HIV Gag-p24 to determine immunogen-specific T-cell responses and to assess functional properties including functional avidity and cross-reactivity in 25 HIV-1 controllers and 25 non-controllers without protective HLA class I alleles. Our data challenge the common belief that Gag-specific T cell responses dominate the virus-specific immunity exclusively in HIV-1 controllers as both groups mounted responses of comparable breadths and magnitudes against the p24 sequence. However, responses in controllers reacted to lower antigen concentrations and recognized more epitope variants than responses in non-controllers. These cross-sectional data, largely independent of particular HLA genetics and generated using direct ex-vivo samples thus identify T cell responses of high functional avidity and with broad variant reactivity as potential functional immune correlates of relative HIV control.


PLOS Computational Biology | 2007

Coping with Viral Diversity in HIV Vaccine Design

David C. Nickle; Morgane Rolland; Mark A. Jensen; Sergei L. Kosakovsky Pond; Wenjie Deng; Mark Seligman; David Heckerman; James I. Mullins; Nebojsa Jojic

The ability of human immunodeficiency virus type 1 (HIV-1) to develop high levels of genetic diversity, and thereby acquire mutations to escape immune pressures, contributes to the difficulties in producing a vaccine. Possibly no single HIV-1 sequence can induce sufficiently broad immunity to protect against a wide variety of infectious strains, or block mutational escape pathways available to the virus after infection. The authors describe the generation of HIV-1 immunogens that minimizes the phylogenetic distance of viral strains throughout the known viral population (the center of tree [COT]) and then extend the COT immunogen by addition of a composite sequence that includes high-frequency variable sites preserved in their native contexts. The resulting COT+ antigens compress the variation found in many independent HIV-1 isolates into lengths suitable for vaccine immunogens. It is possible to capture 62% of the variation found in the Nef protein and 82% of the variation in the Gag protein into immunogens of three gene lengths. The authors put forward immunogen designs that maximize representation of the diverse antigenic features present in a spectrum of HIV-1 strains. These immunogens should elicit immune responses against high-frequency viral strains as well as against most mutant forms of the virus.


The Journal of Infectious Diseases | 2010

Viral evolution and escape during acute HIV-1 infection.

Christian L. Boutwell; Morgane Rolland; Joshua T. Herbeck; James I. Mullins; Todd M. Allen

The extensive genetic diversity of human immunodeficiency virus type 1 (HIV-1) presents a significant barrier to the development of an effective and durable HIV vaccine. This variability not only makes it difficult to identify the targets against which immune responses should be directed, but it also confers on the virus the capacity for rapid escape from effective immune responses. Here, we describe recent investigations of the genetic diversity of HIV-1 at transmission and of the evolution of the virus as it adapts to the host immune environment during the acute phase of HIV-1 infection. These studies increase our understanding of the virology of the earliest stages of HIV-1 infection and provide critical insights into the mechanisms underlying viral replication and immune control of diverse HIV-1 strains. Such knowledge will inform the design of smarter, more effective vaccines capable of inducing immune control of HIV-1.

Collaboration


Dive into the Morgane Rolland's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nelson L. Michael

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Sodsai Tovanabutra

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Wenjie Deng

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Jerome H. Kim

International Vaccine Institute

View shared research outputs
Top Co-Authors

Avatar

Merlin L. Robb

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Brandon Maust

University of Washington

View shared research outputs
Top Co-Authors

Avatar

Eric Sanders-Buell

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Allan C. deCamp

Fred Hutchinson Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge