ÂÒÂ×Ðã

XClose

Institute of Immunity and Transplantation

Home
Menu

Protective and Dysfunctional Antiviral Immunity

We focus on viruses that are major global health challenges. As part of this, we analyse how innate and adaptive immune responses differ in people with efficient versus poor virus control.

Our work

The immune system has the capacity to mount protective immunity to acute and chronic virus infections but when such responses fail, they can instead contribute to the resulting disease pathology. Careful dissection of the molecular basis of protective and harmful immunity using human samples, in addition to appropriate model systems, is essential to tailor novel therapies.

We research both innate and adaptive immune responses, analysing how these differ in those with efficient versus poor virus control. We take advantage of our close links with the Royal Free London to access tissue samples, allowing us to study organ-specific immunity at the sites of infection.

Projects

Capacity of T cells to abort SARS-CoV-2 infection

We have identified an association between memory T cells targeting the conserved replication-transcription complex of SARS-CoV-2 and early viral control without seroconversion (ie abortive infection). We are now characterising these T cells to identify which qualities and specificities confer protection.

Lead Investigators

  • Dr Leo Swadling (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)
  • Professor Mala Maini (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)

Collaborators

  • Professor Mahdad Noursadeghi (Infection, ÂÒÂ×Ðã)
  • Professor Antonio Bertoletti (Duke-NUS, Singapore)

Funding

  • Rosetrees Trust & Pears Foundation – Advancement Fellowship
  • MRC Consortium funding
Harnessing cross-protective airway-resident immunity against SARS-CoV-2 variants

We identified tissue-resident T cells in pre-pandemic human airway samples able to cross-recognise SARS-CoV-2, optimally situated to abort infection through frontline immune surveillance. We are examining persistence of these sentinel mucosal responses following SARS-CoV-2 infection and/or vaccination in UK and Malawi cohorts +/- HIV infection. We will investigate whether airway T and B cells have superior potential for transmission-blocking pan-coronavirus reactivity through enhanced cross-reactivity +/- more rapid/potent antiviral function.

Lead Investigator

  • Professor Mala Maini (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)

Network Collaborators

  • Dr Elena Mitsi (University of Oxford)
  • Dr Daniela Ferreira (University of Oxford)
  • Dr Kondwani Jambo (Liverpool School of Tropical Medicine and Malawi)

Collaborators

  • Professor Tao Dong (University of Oxford)
  • Professor Elma Tchilian (Pirbright Institute)
  • Dr Laura McCoy (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)

Funding

  • Wellcome Covid-19 Variants Network award
  • Wellcome Investigator award
Profile of hepatic HBV-specific T cells for functional cure

We are characterising adaptations of ‘gold-standard’ HBV-specific and global liver-resident T cells surviving and maintaining control in naturally resolved infection and testing immunotherapeutic strategies to recapitulate their functional hallmarks.Ìý

Lead Investigator

  • Professor Mala Maini (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)

Collaborators

  • Dr Leo Swadling (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)
  • Prof Muzlifah Haniffa (University of Newcastle and Sanger Institute)
  • Professor Xavier Forns (Hospital Clínic de Barcelona)
  • Liver surgical team and Tissue Access for Patient Benefit (Royal Free Hospital)

Funding

  • Wellcome Investigator award
Delineation of the TCR signature of HBV control

We are identifying protective TCR sequences from the site of hepatitis-B virus (HBV) infection, the liver, to increase our understanding of immune regulation of this virus and to develop next generation TCR gene therapies.

Lead Investigator

  • Dr Leo Swadling (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)

Collaborators

  • Professor Xavier Forns (Hospital Clínic de Barcelona)
  • Professor Mala Maini (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)

Funding

  • Medical Research Foundation
B cell dysfunction in HIV infection

We are investigating whether alterations in B cell phenotypes explain why only rare individuals are able to make broadly neutralising antibodies against HIV. We are particularly interested in how B cell activation differences are related to antibody affinities.

Lead Investigator

  • Dr Laura McCoy (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)

Collaborators

  • Dr Marit van Gils (University of Amsterdam)
  • Professor Aine McKnight (Queen Mary, University of London)
  • Professor Ravindra Gupta (University of Cambridge)
  • Professor Rachel McKendry (London Nanotechnology Centre)
  • Professor Peter Cherepanov (The Francis Crick Institute, London)
  • Professor Menna Clatworthy (University of Cambridge)

Funding

  • European Research Council
HIV-1 latency

We are investigating how HIV-1 latency is established or reversed. We study how the three-dimensional organization of chromatin in lymphocytes affects the establishment of HIV-1 latency and how HIV-1 gene expression is controlled in specific T helper cell subtypes.Ìý

Lead Investigator

  • Professor Ariberto Fassati (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)

Collaborators

  • Professor Eric Schirmer (Institute of Cell Biology, University of Edinburgh)
  • Professor Petronela Ancuta (CHUM, University of Montreal, Canada).

Funding

  • Medical Research Council
  • Canadian Institute of Health Research
  • Government of Oman
  • Government of Saudi Arabia
Mechanisms of lifelong persistence of human cytomegalovirus

We are dissecting the host and viral mechanisms responsible for controlling latency and reactivation of human cytomegalovirus (HCMV) - a pathogen important for disease in transplant patient populations. Key questions include defining the molecular determinants that regulate viral infection, alongside understanding the immunological basis for control in healthy people. We aim to harness this knowledge to develop strategies to protect our immune-suppressed transplant patients.

Lead Investigator

  • Dr Matthew Reeves (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)

Collaborators

  • Professor Judy Breuer (Institute of Child Health, ÂÒÂ×Ðã)
  • Professor Reza Motallabzadeh (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)
  • Dr. Mark Harber (Royal Free Hospital, ÂÒÂ×Ðã)
  • Professor Mark Wills (University of Cambridge)
  • Professor Richard Stanton (University of Cardiff)

Funding

  • Medical Research Council
  • Wellcome Trust
  • BBSRC
  • Royal Free Charity
  • Qatari Government (Defence Attache)
Tissue resident immunity and the control of human cytomegalovirus replication

Human cytomegalovirus (HCMV) replicates in tissue to cause disease. Understanding the role of tissue resident immunity for the control of viral replication is important for the design of future immunological interventions. We have recently identified that differences in donor-derived immune responses within the transplant organ are a potential correlate of outcome post-transplant in the recipient. The goal now is to understand the role of individual immune functions and why they are protective when enriched for in the transplanted organ.

Lead Investigator

  • Dr Matthew Reeves (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)

Collaborators

  • Professor Reza Motallabzadeh (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)
  • Dr Victoria Male (Imperial College London)
  • Professor Mark Wills (University of Cambridge)

Funding

  • Medical Research Council
  • Kidney Research UK
  • Rosetrees
Polyomavirus disease in renal transplant patients

BK polyoma virus (BKpV) is a major cause of disease in renal transplant patients. Relieving immune suppression is the only mechanism of viral control but leads to graft rejection. We are currently using samples from our patient cohorts to identify immunological correlates of protection with a specific focus on broadly neutralising antibody responses that protect from infection with multiple BKpV genotypes.

Lead Investigator

  • Dr Matthew Reeves (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)

Collaborators

  • Professor Alan Salama, Department of Renal Medicine, ÂÒÂ×Ðã)
  • Dr Stephanie Chong (Department of Renal Medicine, ÂÒÂ×Ðã)
  • Professor Reza Motallabzadeh (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)
  • Dr Laura McCoy (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)

Funding

  • Kidney Research UK
  • ¸é´Ç²õ±ð³Ù°ù±ð±ð²õÌý
  • Royal Free Charity
  • St Peter's Trust
Evolution of NK cell responses during acute HIV infection with distinct viral subtypes

This project aims to identify the innate immune mechanisms involved in the initial response to HIV infection, including identification of NK cell subsets with adaptive/‘memory’ features that can be induced by novel vaccination strategies.

Lead Investigator

  • Dr Dimitra Peppa (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)

Collaborators

  • Professor Sarah Rowland Jones (University of Oxford)
  • Professor Eduard Sanders (Kilifi, Kenya)

Funding

  • BHIVA
  • Saudi Ministry of Education Prize scholar (D.Phil. Student)
The prevalence and activity of anti-interferon auto-antibodies in people living with chronic virus infections

Type 1 interferon (T1IFN) signalling is critical in immune responses to viruses. Auto-antibodies (Abs) that bind and neutralise T1IFN are strongly associated with poor outcomes during SARS-CoV-2, influenza and West Nile Virus disease. We will measure the full spectrum anti-IFN auto-Abs (across T1IFN, T2IFN and T3IFN sub-types) in people living with chronic virus infection, including HIV and chronic hepatitis B virus, and characterise associations with key virological and clinical outcomes.
Ìý
Lead Investigator

  • Dr Douglas Fink (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)

Collaborators

  • Professor Mala Maini (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)
  • Dr Laura McCoy (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)
  • Dr Dimitra Peppa (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)
  • Dr Upkar Gil (Queen Mary, University of London)

Funding

  • British HIV Association
  • ÂÒÂ×Ðã Therapeutic Acceleration Support Fund
Mucosal Immunity in human Coronavirus Challenge

Coronaviruses typically infect people through cells lining their nose, throat and lungs. Mucosal immunity generated at these surfaces is highly specialised and very different to immune responses in the circulation. Boosting mucosal immunity could be the key to developing vaccines that can block viruses from being able to spread from one person to another. The five-year Mucosal Immunity in human Coronavirus Challenge (MusiCC) project will be led by Imperial College London and involve collaborators across the UK, Europe, Singapore and USA. The consortium will develop human challenge models for coronavirus trials and mechanistic studies of mucosal immunity. The trials will test potential mucosal vaccine candidates against beta-coronaviruses. ÂÒÂ×Ðã and the Royal Free Hospital will deliver clinical and laboratory work packages within the consortium.
Ìý
Chief Investigator

  • Professor Christopher Chiu (Imperial College London)

Principal Investigator

  • Dr Douglas Fink (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)

Collaborators

  • Professor Rob Heyderman (ÂÒÂ×Ðã)
  • Professor Mahdad Noursadeghi (ÂÒÂ×Ðã)
  • Professor Helen McShane (University of Oxford)

Funding

  • Coalition for Epidemic Preparedness Innovations (CEPI)Ìý
  • Horizon Europe
Circadian regulation of viral infection and immune responser

The circadian clock is a crucial regulator of immunity, influencing how the body defends against pathogens. Our research aims to understand how the circadian system regulates immune responses to viral infections, with a specific focus on the hepatitis B virus. This will allow us to optimise the time-of-day for vaccine administration to improve efficacy. Additionally, our work involves characterising the communication between immune cells and the liver stroma in the context of the circadian clock and investigating how infections may alter this communication.

Lead Investigator

  • Dr Alan Xiaodong Zhuang (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)

Collaborators

  • Professor Russell Foster (University of Oxford)
  • Professor Mala Maini (Institute of Immunity & Transplantation, ÂÒÂ×Ðã)

Funding

  • Medical Research Council

Our Experts

Leo Swadling


Pears &ÌýRosetrees Fellow

Mala Maini


Viral Immunity

Dr Laura McCoy


Associate Professor

Ariberto Fassati


Cellular & Molecular Virology

Matt Reeves


Associate Professor

Dimitra Peppa


HIV Immunology

Douglas Fink


Principal Clinical Fellow

Alan Zhuang


Infection &ÌýImmunity


Funding and Partnerships

Logo for the Rosetrees Trust

Logo for the Pears Foundation

Logo for the Medical Research Foundation

logo for welcome trust uk

Logo for the Royal Free Charity

Logo for medical research council

Logo for Canadian Institutes for Health Research

Logo for the State of Oman

Vector logo for the state of Saudi Arabia

logo for bbsrc

logo etc

Logo for the Qatar State

Logo for the British HIV Association

Logo for the Saudi Arabia Ministry of Education

Logo for the Coalition for Epidemic Preparedness Innovations (CEPI)

Logo for Kidney Research UK

Logo for St Peter's Trust (Royal Free)

Logo for Horizon 2020

Ìý

Selected Publications

  1. Sun B, da Costa KAS, Alrubayyi A ...ÌýMaini MK, Bashford-Rogers R, Gill US, Peppa D (2024). . Hepatology. 2024 Sep 1;80(3): 649-663.

  2. Fink DL, Idilli O, Shields A, et alÌý(2024). . J Clin Immunol. 2024 Jul 11;44(7): 161.

  3. Thomas P, Rees-Spear C, Griffith S ...ÌýMcCoy LEÌý(2024). . iScience. 2024 Mar 11;27(4): 109495.

  4. Moreno-Cubero E, Alrubayyi A, Balint SÌý...ÌýPeppa D (2024). . JCI Insight. 2024 Jan 16;9(4): e173099.

  5. Murray MJ, Bradley E, Ng Y ...ÌýReeves MB (2023). . J Biol Chem. 2023 Jun;299(6): 104727.

  6. Borrmann H, Ulkar G, Kliszczak AE ...ÌýZhuang X, McKeating JA (2023). . iScience. 2023 May 29;26(7): 107007.

  7. Forrest C, Chase TJG, Cuff AO ...ÌýReeves M (2023). . Nat Commun. 2023 Mar 14;14(1): 1409.

  8. Touizer E, Alrubayyi A, Ford R ...ÌýPeppa D, McCoy LE (2023). . iScience. 2023 Jan 20;26(1): 105862.

  9. Fink DL, Callaby H, Luintel A,Ìýet al (2023). . Lancet Infect Dis. 2023 May;23(5): 589-597.

  10. Alrubayyi A, Moreno-Cubero E, Hameiri-Bowen D ...ÌýPeppa D (2022). . Front Immunol. 2022 Jul 5;13: 908697.

  1. Diniz MO, Schurich A, Chinnakannan SK ...ÌýMaini MK (2022). . Sci Transl Med. 2022 Apr 13;14(640): eabi4670.

  2. Wiche Salinas TR, Zhang Y, Sarnello D ...ÌýFassati A (2021). . Proc Natl Acad Sci USA. 2021 Nov 30;118(48): e2105927118.

  3. Zhuang X, Tsukuda S, Wrensch F, et al (2021). . iScience. 2021 Oct 22;24(10): 103144.

  4. Rees-Spear C, Muir L, Griffith SAÌý...ÌýMcCoy LE (2021). . Cell Rep. 2021 Mar 23;34(12): 108890.

  5. Zhuang X, Forde D, Tsukuda S, et alÌý(2021). . Nat Commun. 2021 Mar 12;12(1): 1658.

  6. Swadling L, Pallett LJ, Diniz MO ...ÌýMaini MK (2020). . Cell Rep. 2020 Jan 21;30(3): 687-698.e6.

  7. Monit C, Morris ER, Ruis C ...ÌýFassati A, Goldstein RA (2019). . Proc Natl Acad Sci USA. 2019 Sep 10;116(37): 18647-18654.

  8. Dupont L, Du L, Poulter M, Choi S, McIntosh M, Reeves MB (2019). . J Biol Chem. 2019 Aug 30;294(35): 12901-12910.

  9. Burton AR, Pallett LJ, McCoy LE, Suveizdyte K, Amin OE, Swadling LÌý...ÌýMaini MK (2018). . J Clin Invest. 2018 Oct 1;128(10): 4588-4603.

  10. Zhyvoloup A, Melamed A, Anderson I ...ÌýFassati A (2017). . PLoS Pathog. 2017 Jul 20;13(7): e1006460.