Professor Luk Vandenberghe

Man in white shirt holding up virus model

Research in my laboratory pursues the development of the science and technology needed to support the broad applications of therapeutic gene transfer for unmet disease. It aims to do so by pursuing questions relevant to (a) preclinical programs that aim to translate gene therapies to Phase I/II studies, (b) the development of gene transfer technologies, and (c) the study of fundamental mechanism pertinent to relevant disease or somatic gene transfer biology.

I direct the Grousbeck Gene Therapy Center at Mass Eye and Ear, part of Mass General Brigham and I am the Grousbeck Family Chair in Gene Therapy as Associate Professor at Harvard Medical School. The Center seeks to fuel the therapeutic gene transfer efforts on the Harvard campus by centralizing key resources such as vector production and analytics, as well as providing key expertise. We further pride ourselves in the training of talent for the growing gene therapy field and have over a dozen of trainees at all levels now leading successful careers in academia and industry. I am keen to develop novel pathways for academic innovation to be furthered in human studies; previously I have founded a non-profit biotech (Odylia), 4 gene therapy companies (GenSight, Akouos, Affinia, and Albamunity), and collaborate on the plan for Harvard University to develop a GMP manufacturing side for cell and gene therapy. Our work has further led to technologies now being used in over 12 clinical trials and 1 product (Zolgensma).

As part of the CureHeart project, we seek to develop cardiac specific AAV-based gene transfer technologies that overcome limitation with the current state-of-the-art technologies. In addition, through the Gene Transfer Vector Core facility that I direct, my group will provide standardized AAV reagents to other partners within the consortium, as well as translational gene therapy guidance.

Find out more about Professor Vandenberghe's work. 

Publications

Therapeutic Gene transfer and Translational gene therapy

Our work has focused extensively at discovery and characterization of vector reagents to understand and improve safety and efficacy of therapeutic gene therapy. A major focus of our work preclinically has been on targeting the central nervous system and neurosensory organs. In addition, our work extensively described and provides mechanism to vector-host interactions that determined efficacy and safety. We identified several highly neurotropic novel AAV serotypes, evaluated immunological response to the vector and transgene antigens upon administration, and did so in small and larger animal models.

Specifically, our work has evaluated passive and active transport upon stereotactic injection, tropism in the retina following various routes of administration, and more recently vector targeting in the cochlea. Lastly, our independent work has recently led to a novel set of gene transfer reagents based on in silico design of AAV that demonstrate high liver tropism and distinct immunological reactivity as compared to existing vectors. Translational aspects of our work extended to non-neuronal gene therapy targets including liver and muscle, most recently with an application for genome editing in Duchenne Muscular Dystrophy. Vectors studied and/or developed by us now support clinical trials for Hemophilia B and Spinal Muscular Atrophy I.

  • Vandenberghe LH, Bell P, Maguire AM, Cearley CN, Xiao R, Calcedo R, Wang L, Castle MJ, Maguire AC, Grant R, Wolfe JH, Wilson JM, Bennett J. 2011. Dosage thresholds for AAV2 and AAV8 photoreceptor gene therapy in monkey. Sci Transl Med 3:88ra54. doi: 10.1126/scitranslmed.3002103.
  • Landegger, L. D., Pan, B., Askew, C., Wassmer, S. J., Gluck, S. D., Galvin, A., Taylor, R., Forge, A., Stankovic, K. M., Holt, J. R., & Vandenberghe, L. H. (2017). A synthetic AAV vector enables safe and efficient gene transfer to the mammalian inner ear. Nat Biotechnol. doi:10.1038/nbt.3781
  • Hudry, E. & Vandenberghe, L. H. Therapeutic AAV Gene Transfer to the Nervous System: A Clinical Reality. Neuron 101, 839-862, (2019). doi:10.1016/j.neuron.2019.02.017

Immunobiology of viral gene transfer

Historically, immunity has arguably been largest obstacle toward safe in-vivo gene therapy with potential immunotoxicity arising from both vector and transgene antigens. Additionally, many viruses used for gene therapy, are prevalent in human populations resulting in pre-existing immunity (PEI) toward vector capsid antigens that result in neutralization and blocking of gene transfer and potential immune sequelae. Our studies have characterized PEI in global seroprevalence screen that is cited as the most comprehensive study in this regard, and influencing vector decisions for clinical translation. Furthermore, we have modeled in animals a component of a T-cell activation toward the AAV capsid and identified the AAV2 heparin binding motif to be a key determinant in this immunological observation which led to the halting of a hemophilia B AAV2 Phase I/II trial. AAV8, which we show was not able to activate these T-cell subset, was proposed as an alternative vector which was later used to develop a Hemophilia B gene therapy with lasting efficacy (albeit with some remaining idiosyncratic immunity issues that remain to be resolved). We furthermore have characterized mechanistically (mouse) and descriptively (mouse and nonhuman primate) cytotoxic immunity toward non-self antigens in gene transfer models. We believe these data have highlighted the margins within current AAV technology can operate safely, and provided some initial solutions toward overcoming some of the remaining challenging hurdles around immunity in an in vivo gene therapy context.

  • Vandenberghe LH*, Wang L*, Somanathan S, et al. Heparin binding directs activation of T cells against adeno-associated virus serotype 2 capsid. Nat Med 2006;12:967-71. doi: 10.1038/nm1445.
  • Vandenberghe LH*, Calcedo R*, Gao G, Lin J, Wilson JM. Worldwide epidemiology of neutralizing antibodies to adeno-associated viruses. J Infect Dis 2009;199:381-90. doi: 10.1086/595830.

Viral Gene Transfer Vector Discovery and Development

Gene delivery is key rate limiting step in therapeutic applications. The gene transfer vector capsid or envelope provides the primary interface with the host that determines most aspects that govern the safety and efficacy of gene therapy applications (e.g. tropism, vector immunobiology, etc.). Our efforts have focused on the discovery of novel adeno-associated virus and adenoviral vector reagents. In one effort, through biomining nonhuman and human primate tissue samples we identified several novel clades of AAV that we subsequently developed and characterized as gene therapy vectors. Many of these vectors have been extensively used in the broad gene therapy research community. One of these vectors, AAV9, now entered clinical trials (led by others) for treatment of spinal muscular atrophy and was shown by us to demonstrate unique transduction properties for cone photoreceptor targeting in the non-human primate retina. This work led to 8 patents, some of which are licensed for therapeutic development. In our most recent work, we propose the use of in silico derived AAVs based on maximum-likelihood ancestral sequence reconstruction methodologies in order to develop synthetic particles that are disrupted in immunodominant epitopes that govern pre-existing immunity to AAV, a primary hurdle in clinical gene therapy.

  • Vandenberghe, LH, Gao, G, Alvira, MR, Lu, Y, Calcedo, R, Zhou, X, and Wilson, JM (2004). Clades of Adeno-associated viruses are widely disseminated in human tissues. J Virol 78: 6381-6388. doi: 10.1128/JVI.78.12.6381-6388.2004.
  • Vandenberghe LH, Xiao R, Lock M, Lin J, Korn M, Wilson JM. Efficient serotype-dependent release of functional vector into the culture medium during adeno-associated virus manufacturing. Hum Gene Ther 2010;21:1251-7. doi: 10.1089/hum.2010.107.
  • Zinn, E., Pacouret, S., Khaychuk, V., Turunen, H.T., Carvalho, L.S., Andres-Mateos, E., Shah, S., Shelke, R., Maurer, A.C., Plovie, E., et al. (2015). In Silico Reconstruction of the Viral Evolutionary Lineage Yields a Potent Gene Therapy Vector. Cell Rep 12, 1056-1068. doi: 10.1016/j.celrep.2015.07.019.

Structure-function studies of AAV

AAV is not only one of the smallest and simplest mammalian viruses, it has also captivated the world of gene therapy as AAV, with remarkable efficiency and safety, is able to deliver and transduce genetic cargo into therapeutic target tissues such as the retina, liver, and CNS. AAVs come in many flavors that were originally distinguished based on serology, but now are more and more phylogenetically and structurally defined. Different AAVs demonstrate very distinct phenotypes (tropism, receptor use, host response, assembly, etc.) as a wild type virus or replication-defective vector. We are interested in functionally and structurally understanding the mechanism of these stark differences from the perspective of virus as well as host. One avenue towards this goal is to study the evolutionary biology of this virus, and use mathematical, statistical, and systems approaches in combination with empirical and molecular methods to shed light on the structure-function relationship of these small, yet intricately complex infectious protein assemblies. Within the relatively small genetic AAV space, we have observed stark phenotypic differences affecting tropism for retina, liver, muscle, and conducting airway, and the ability of AAV to egress from the cell during viral vector production, immunological properties. These findings have allowed us to develop a limited predictive structure-function model of AAV that allowed us to improve vector reagents in their gene transfer efficiency by site-directed mutagenesis, including AAVrh.32.33, a functionally unique AAV which we collaboratively developed a X-ray crystallography model for. More recent work has developed fully in silico methods to generate novel AAVs with desirable properties. This modeling effort is being guided by a deeper understanding of the evolutionary history of AAV. These studies ultimately aim to develop a blueprint of AAV that allows for tailoring this therapeutic delivery vector for improved safety and efficacy in the clinic.

  • Dudek, A.M., Zabaleta, N., Zinn, E., Pillay, S., Zengel, J., Porter, C., Franceschini, J.S., Estelien, R., Carette, J.E., Zhou, G.L., et al. (2020). GPR108 Is a Highly Conserved AAV Entry Factor. Mol Ther 28, 367-381. doi: 10.1016/j.ymthe.2019.11.005.
  • Vandenberghe LH, Breous E, Nam HJ, et al. Naturally occurring singleton residues in AAV capsid impact vector performance and illustrate structural constraints. Gene Ther 2009;16:1416-28. doi: 10.1038/gt.2009.101.

A complete published bibliography is also available.