Host-Directed Therapies

Pipeline We study the host-pathogen interaction to improve our understanding regarding how the host immune system responds to M.tb infection and how this is manipulated by M.tb to propagate infection, to help identify targets for host-directed therapy (HDT). There are many benefits to HDT, including that HDTs:

  • are expected to work against both drug susceptible and resistant TB
  • provide a treatment option that the pathogen is unlikely to develop resistance to
  • shorten treatment duration
  • boost the immune response
  • ameliorate pathology associated with severe disease.

HDTs could be used in conjunction with antibiotics and could become an important tool in the fight against infectious diseases like TB. Our pipeline for testing HDTs for TB consists of first testing efficacy in human and murine macrophages, then testing those compounds in a more complex human granuloma model that we developed, to identify compounds that can penetrate and retain activity in macrophages and the unique granuloma environment.

Selected Publications

  • Arnett, Eusondia, Susanta Pahari, Chrissy M Leopold Wager, Elizabeth Hernandez, Jordan R Bonifacio, Miranda Lumbreras, Charles Renshaw, Maria J Montoya, Joseph T Opferman, and Larry S Schlesinger. (2023) 2023. “Combination of MCL-1 and BCL-2 Inhibitors Is a Promising Approach for a Host-Directed Therapy for Tuberculosis”. Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie 168: 115738. https://doi.org/10.1016/j.biopha.2023.115738.

    Tuberculosis (TB) accounts for 1.6 million deaths annually and over 25% of deaths due to antimicrobial resistance. Mycobacterium tuberculosis (M.tb) drives MCL-1 expression (family member of anti-apoptotic BCL-2 proteins) to limit apoptosis and grow intracellularly in human macrophages. The feasibility of re-purposing specific MCL-1 and BCL-2 inhibitors to limit M.tb growth, using inhibitors that are in clinical trials and FDA-approved for cancer treatment has not be tested previously. We show that specifically inhibiting MCL-1 and BCL-2 induces apoptosis of M.tb-infected macrophages, and markedly reduces M.tb growth in human and murine macrophages, and in a pre-clinical model of human granulomas. MCL-1 and BCL-2 inhibitors limit growth of drug resistant and susceptible M.tb in macrophages and act in additive fashion with the antibiotics isoniazid and rifampicin. This exciting work uncovers targeting the intrinsic apoptosis pathway as a promising approach for TB host-directed therapy. Since safety and activity studies are underway in cancer clinics for MCL-1 and BCL-2 inhibitors, we expect that re-purposing them for TB treatment should translate more readily and rapidly to the clinic. Thus, the work supports further development of this host-directed therapy approach to augment current TB treatment.

  • Wager, Chrissy M Leopold, Jordan R Bonifacio, Jan Simper, Adrian A Naoun, Eusondia Arnett, and Larry S Schlesinger. (2023) 2023. “Activation of Transcription Factor CREB in Human Macrophages by Mycobacterium Tuberculosis Promotes Bacterial Survival, Reduces NF-KB Nuclear Transit and Limits Phagolysosome Fusion by Reduced Necroptotic Signaling”. PLoS Pathogens 19 (3): e1011297. https://doi.org/10.1371/journal.ppat.1011297.

    Macrophages are a first line of defense against pathogens. However, certain invading microbes modify macrophage responses to promote their own survival and growth. Mycobacterium tuberculosis (M.tb) is a human-adapted intracellular pathogen that exploits macrophages as an intracellular niche. It was previously reported that M.tb rapidly activates cAMP Response Element Binding Protein (CREB), a transcription factor that regulates diverse cellular responses in macrophages. However, the mechanism(s) underlying CREB activation and its downstream roles in human macrophage responses to M.tb are largely unknown. Herein we determined that M.tb-induced CREB activation is dependent on signaling through MAPK p38 in human monocyte-derived macrophages (MDMs). Using a CREB-specific inhibitor, we determined that M.tb-induced CREB activation leads to expression of immediate early genes including COX2, MCL-1, CCL8 and c-FOS, as well as inhibition of NF-kB p65 nuclear localization. These early CREB-mediated signaling events predicted that CREB inhibition would lead to enhanced macrophage control of M.tb growth, which we observed over days in culture. CREB inhibition also led to phosphorylation of RIPK3 and MLKL, hallmarks of necroptosis. However, this was unaccompanied by cell death at the time points tested. Instead, bacterial control corresponded with increased colocalization of M.tb with the late endosome/lysosome marker LAMP-1. Increased phagolysosomal fusion detected during CREB inhibition was dependent on RIPK3-induced pMLKL, indicating that M.tb-induced CREB signaling limits phagolysosomal fusion through inhibition of the necroptotic signaling pathway. Altogether, our data show that M.tb induces CREB activation in human macrophages early post-infection to create an environment conducive to bacterial growth. Targeting certain aspects of the CREB-induced signaling pathway may represent an innovative approach for development of host-directed therapeutics to combat TB.