PhD Projects for 2025 Entry
The following are industry-linked 4-year PhD studentship projects that we are offering through the Centre for Doctoral Training for a start in September 2025.
This is not an exhaustive list of all projects and these are subject to modification depending on final contract agreements and student recruitment.
Engineering of adipose bone marrow niches of acute myeloid leukemia for safety and efficacy testing
Supervisors: Prof Julien Gautrot, Dr Paulo Gallipoli
Industry Partner: Astra Zeneca
Acute myeloid leukaemia (AML) affects an increasing number of patients worldwide (for >60 year old population, >60k/year; 10-15% survival rate). AML is aggressive and typically results in the hyper-proliferation of immature myeloblasts in the bone marrow, resulting in impaired haematopoiesis. The adipose content of the BM niche is associated both with the resistance of AML cells to therapies and the resurgence of the disease. However, the impact of the adipose BM microenvironment on chemoresistance and how it provides a niche for AML cells to persist and resurge is unknown. In this project, the impact of microenvironment engineering on AML cell resistance and resurgence will be investigated.
- The design and characterisation of biomimetic bone marrow microenvironments will be developed. A range of techniques including scanning electron microscopy, confocal microscopy and nanoindentation will be used to compare the resulting microenvironments to bone marrow samples.
- The regulation of HSCs and AML cells in the resulting microenvironments will be characterised via confocal microscopy, flow cytometry and RNA seq, applying some of the methods previously developed by our teams.
- The response of HSC and AML cells to a panel of therapeutics known to be associated with tumour resurgence will be investigated via dose responses and follow up tumour resurgence assays (with RNA seq of the final time points for comparison to patient data).
The project will result in a novel assay enabling the systematic testing of therapeutics safety and efficacy in the context of AML resistance.
Development of a vascularised muscle-tendon inflammation-on-a-chip model
Supervisors: Prof Hazel Screen, Prof Martin Knight.
Industry Partner: UCB Pharma
Myositis is a chronic, progressive and debilitating muscle wasting condition, which causes muscle to become thin and weak. The causes are unknown, but it is thought to be a group of systemic autoimmune diseases, driven by muscle inflammation. With no cure or effective therapies, there is a need for better in vitro models of myositis, with which to explore disease processes and possible treatments.
- This project is a collaboration with the pharmaceutical company UCB, with the overall aims to design and build a vascularised muscle-tendon chip and use it to explore the drivers of myositis.
- The project will address the design of appropriate biomaterials to recapitulate muscle and tendon niche environments, and then explore the use of gradients in biomaterial stiffness or composition to capture the muscle-tendon interface. It will integrate these materials into an overall organ-chip model, bringing a vasculature, to provide a vascularised muscle-tendon model.
- Once the model has been validated, it will be used to explore myositis. Myositis patient derived cells will be compared with healthy cells, using the model to subject the cells to different physiochemical drivers such as mechanical strain and inflammation, to explore the mechanobiology driving myositis.
Collectively, this project will not only develop new myositis in vitro models for the community but use these to provide novel insights into the drivers of this debilitating disease. The student will be part of a large multi-disciplinary organ-chip research group and will be supported by 3 post docs working on a new organ-chip grant in this area of organ-chip technology (EPSRC grant: Organ-chips for accelerated deployment of new medicines)
An organ-chip model of inflammation in the canine cartilage-synovium interface
Supervisors: Dr Stefaan Verbruggen, Prof Martin Knight.
Industry Partner: Zoetis
As part of the standard preclinical drug pipeline, candidate therapies developed for humans are often tested for safety and efficacy on rodent or primate models before moving into Phase 1 clinical trials. Many of these animal tests, including some specifically in feline or canine models, provide key data for development of veterinary medicines for companion animals such as cats and dogs. However, with the movement away from animal use in science and towards microphysiological systems using human cells, the development of treatments specifically for animals may stall.
- This presents both a challenge, but also an opportunity, as there is potential to develop species-specific organ-on-a-chip models to tackle a range of conditions faced by companion animals.
- One particularly widespread condition seen in the veterinary clinic is canine osteoarthritis (OA), with most breads of dog suffering from some degree of this disease in old age. Canine joint physiology is significantly different from humans, and therefore treatments developed for humans are unlikely to translate to dogs.
- Therefore, the aim of this project is to develop an organ-on-a-chip model of the canine cartilage-synovium interface, using it to characterise the degradation and inflammation that occurs in OA. Organ-chip technology is particularly relevant for this condition, as the joint represents a highly mechanically active tissue.
Once developed, this chip will be used to investigate the cascades initiated by disease relevant inciters of chondrocyte damage, such as pressure, shear stress, genetic mutation, and physiological levels of various inflammatory cytokines, providing a novel tool to develop veterinary medicines for musculoskeletal conditions in dogs.
Effect of mechanical environment on immune cell invasion within an organ-chip
Supervisors: Prof Martin Knight, Prof Hazel Screen
Industry Partner: Emulate
Organ-on-a-chip technology has the potential to transform both fundamental hypothesis driven research and pre-clinical therapeutic testing. The incorporation of circulatory immune cells into organ-chip models is important for accurately replicating inflammatory conditions associated with a wide range of diseases. These include inflammation in tissues such as arteries leading to atherosclerosis; cartilage and synovium linked to osteoarthritis; and various cancers and metastases.
This industry-linked, multidisciplinary project will examine the influence of the physiological and pathological biomechanical environment on immune cell adhesion, invasion and migration within organ-chip models. This will involve analysis of the biological effects of (1) substrate stiffness and porosity associated with the use of extracellular matrix and 3d biomaterials; (2) applied tensile strain to mimic physiological loading; and (3) fluid shear forces to replicate blood flow. The aim of the study is to test the fundamental hypothesis that invasion of immune cells is regulated by biomechanical environment and to develop protocols that maximise immune cell incorporation into organ-chip models through manipulation of biomechanics. Initial studies will utilise THP-1 monocytes, whilst further studies will investigate the behaviour of a variety of immune cells including patient derived cells. Cells will be stimulated with pro-inflammatory cytokines including IL-1β, IL-6, TNFα, and IFNγ.
The project is supported by Emulate Inc., one of the leading organ-chip companies who have a long-standing partnership with Queen Mary via the Centre for Predictive in vitro Models. The work will focus on the use of the Emulate organ-chip platform and will involve training in techniques including cell culture, confocal microscopy, molecular biology, biochemistry, and mechanobiology.
The student will be part of a large multi-disciplinary organ-chip research group and will be supported by 3 post docs working on a new organ-chip grant in this area of organ-chip technology (EPSRC grant: Organ-chips for accelerated deployment of new medicines)
Tissue in a tube bioreactor screening of bioresorbable composites for bone repair
Supervisors: Prof Karin Hing, Prof Simon Rawlinson
Industry Partner: TBC
We are inviting applications for an exciting multidisciplinary project, to showcase the use of a Tissue in-a Tube (TiaT) perfusion bioreactor model of bone healing for use in optimisation of orthobiologic scaffolds to support accelerated bone healing.
This exciting studentship will involve investigation of both the formulation and scaffold structure of a novel orthobiologic composite to maximise a pro-osteogenic response from human mesenchymal stem cells (hMSC) cultured within the TiaT model. The TiaT model represents an important step forward from traditional 2D or static 3D cultures, where mechanical signals associated with interstitial fluid movement are generally ignored. The model is not complicated, and easy to establish in any laboratory. The current barriers to effective use of in vitro testing in biomaterials safety and efficacy testing are the continued dependence on 2D models, and the lack of recognition of the importance of mechanical input influencing paracrine signalling and biological response.
However, now is an ideal time to advance this model and its potential, and this studentship will additionally explore the impact of introducing mechanical loading of the biomaterial scaffolds to the model. Pro-osteogenic responses will be monitored through a combination of histological and immunological analysis of extracellular matrix development, proteomic analysis of markers of hMSC osteogenic differentiation and quantification of cellular soluble mediator release profiles over incubation periods of up to 28 days. Where cells will be seeded within the orthobiologic scaffolds and systematically subjected to physiologically relevant levels of interstitial fluid flow and mechanical loading.
Development of a dual synovial joint and heart on-a-chip model for investigating heart disease in Rheumatoid arthritis and testing new therapeutics
Supervisors: Dr Lucy Norling, Dr Jianmin Chen, Prof Thomas Iskratsch
Industry Partner: SynAct Pharma
This studentship will be in collaboration with SynAct Pharma, a clinical-stage biotechnology company, which develop innovative therapies for inflammatory conditions e.g. Rheumatoid arthritis (RA). Whilst RA itself is not lethal, around half of all premature deaths in RA patients are due to cardiovascular comorbidities, among which non-ischaemic heart failure plays a predominant role. The causes are unknown and pre-clinical research into this condition is hindered by a lack of suitable models. This studentship will address this unmet need by developing an interconnected model of heart and synovial joint on-a-chip to further explore the cross-talk between the different organs in the context of arthritis. Cardiac fibroblasts and cardiomyocytes will be derived from iPSC to mimic the structural components and biomechanical properties of the heart.
We hypothesise that soluble factors released from the inflamed synovium drive cardiac dysfunction by altering stromal cell phenotype and therefore contractility of the heart.
The main objectives will be to:
- Design and optimize organ-chip models that accurately recapitulate key aspects of inflammatory arthritis, including synovial inflammation and explore the impact this has on the heart via interconnected chips.
- Utilise the in vitro model to explore the underlying molecular and cellular mechanisms contributing to arthritis development and heart disease progression.
- Employ the established in vitro model to screen and evaluate new therapeutics for arthritis treatment and cardiac protection.
Overall, the student will develop deep technical bioengineering skills, characterising a new organ-chip model to increase understanding of arthritis progression and emergence of secondary organ damage, enabling clinical translation.
Generation and testing of a cutting-edge organ-on-a-chip technology for polycystic kidney disease
Supervisors: Dr Maria Fragiadaki, Prof Thomas Iskratsch
Industry Partner: Vertex
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a genetic condition that affects over 12 million of people, with no cure, resulting in renal failure. It primarily stems from mutations in Polycystic Kidney Disease 1 (PKD1) gene, triggering over-proliferation, apoptosis and autophagy, causing progressive renal cyst growth. Tolvaptan is used to manage the symptoms of ADPKD, but unfortunately comes with severe side effects, such as liver failure. Generating models to study new therapeutic targets is of paramount importance.
In this project we will develop an advanced organ-on-a-chip model for studying ADPKD, using established cells lines and patient-derived renal tubular epithelial cells. We will incorporate fluid flow, which is the frictional force exerted onto cell to adjust cellular behaviour, and extracellular matrix that mimics the human kidney closely. PKD1 will be genetically modified and assays such as viability, proliferation and autophagy will be performed to assess the efficiency of the newly generating model in replicating key responses to drug stimulation. To begin with the student will use drugs with known effects, such as tolvaptan and JAK inhibitors. Eventually the model will be used to test the effectiveness of novel medicines.
Hence, this project aims to create a sophisticated cutting-edge tool for ADPKD research that can significantly advance our understanding of the disease and accelerate the development of new therapies without relying on animal testing.
Vaginal probiotics for reproductive health: microbiome organ-chip
Supervisors: Dr Tina Chowdhury, Dr Ruairi Robertson, Dr Stefaan Verbruggen
Industry Partner: Calla Lily Clinical Care
Bacterial vaginosis is a vaginal infection that doubles the risk of women to sexually transmitted diseases like HIV and increases the risk of preterm birth affecting the menstrual to menopause life-course. In the UK, around 30% of reproductive aged women are affected by bacterial vaginosis and whilst treated with antibiotics, the infection recurs leading to complications such as infertility, endometriosis and pelvic floor disorders.
Bioengineers at Calla Lily Clinical Care have studied probiotics to treat bacterial vaginosis by delivering live bacteria using a novel, patented device. However, it is not clear how the bacterial formulation works on infection and inflammatory mechanisms and whether the combination device could be used as a clinical intervention to treat women with bacterial vaginosis.
In this project, the PhD research student will work with an inter-disciplinary team of clinicians, microbiome and infection scientists at the Centre for Bioengineering, Blizard Institute, UCLH and Barts Health NHS Trust. Our goal will be to evaluate the effects of anti-microbial and probiotic regimens to treat bacterial vaginosis. You will develop a human vagina microbiome chip model, determine the effect of bacterial combinations on the vaginal mucosal barrier, and test survival and compatibility of probiotic strains to treat bacterial vaginosis using the medical device patented by Calla Lily Clinical Care.
The research student will develop inter-disciplinary skills, learn techniques in 3D organ chip culture, cell/molecular biology, imaging, biochemistry and computational modelling.
Preference will be given to candidates who have an interest in infection and inflammation research with a background in biomedical sciences and are interested in developing a career Women’s Health.
Investigating immune dysfunction in 3D microfluidic models of inflammatory skin disease
Supervisors: Prof John Connelly, Prof Martin Knight
Industry Partner: GSK
The skin is a complex immunologic barrier to the external environment, and replicating the tissue’s intricate immune processes in vitro remains a significant challenge. In collaboration with GSK, this studentship aims to develop a novel immuno-competent model of human skin infection. The project will employ advanced biofabrication methods to engineer microfluidic lymphatics and resident immune populations within 3D human skin equivalents (HSEs), and the system will be validated molecularly and functionally. The specific aims include:
- Engineer microfluidic lymphatic vessels. Fluidic microchannels in the HSE dermal compartment will be fabricated using digital light-based printing methods, lined with lymphatic endothelial cells, and characterised structurally and molecularly by microscopy.
- Engineer spatially-defined resident immune cells. Light-based coupling reactions will be used to pattern cell-instructive ligands for defining myeloid lineages within the dermis, and monocyte fate and function in response to these cues will be analysed by microscopy and flow cytometry.
- Characterise the response to infection. HSEs with lymphatics and resident immune cells will be challenged with a range of antigens. Resident immune cell trafficking through the tissue and lymphatics will be analysed by microscopy, and molecular signatures will be analysed by flow cytometry and transcriptomic profiling.
- Investigate the effects of chronic inflammation. Low-dose cytokine treatment will be used to mimic chronic inflammatory states, such as eczema, and the response of resident immune cells to infection will be analysed as before.
The findings will establish next-generation microfluidic models of immuno-competent skin and provide fundamental insights into human-specific responses to infection.
Developing artery on a chip technology to identify patients with life-threatening heart disease
Supervisors: Prof Paul Evans, Prof Thomas Iskratsch
Industry Partner: Ibidi
This EPSRC-funded PhD studentship offers an exciting opportunity to advance patient care for coronary artery disease (CAD) by developing personalized "vessel-on-a-chip" models to predict accelerated disease progression. CAD, caused by the buildup of arterial plaques, poses significant risks such as unstable angina and heart attacks. However, disease progression varies, with some high-risk patients responding differently to mechanical factors like shear stress. This project aims to pinpoint patient-specific responses to these forces, paving the way for tailored treatments and enhanced patient stratification.
The four-year studentship includes extensive training in cell culture, the use of cutting-edge 2D and 3D vessel-on-a-chip systems, and the analysis of patient-specific flow responses. In the first year, the student will master cell culture and flow techniques and gain clinical exposure in a catheterisation lab. Year two focuses on using Ibidi’s 2D flow systems to study inflammatory and apoptotic responses. During a placement at Ibidi’s industry labs in year three, the student will develop a novel 3D vessel-on-a-chip model. The final year will integrate both 2D and 3D technologies to evaluate patient-specific responses, correlating findings with clinical data.
This project provides a unique blend of academic and industrial experience, giving students hands-on training with a leading industry partner, Ibidi. Aligned with EPSRC's goals in precision medicine and quantitative research, this studentship offers a strong foundation for those seeking careers in translational research and medical technology development
An artery-on-a-chip model to investigate the communication between different arterial cell types
Supervisors: Prof Thomas Iskratsch, Prof Paul Evans
Industry Partner: Qiagen (TBC)
Vascular smooth muscle cells (VSMCs) take a central role in the regulation of vascular tone, but also the ageing and/or disease associated arterial remodelling. In the normal arterial wall, VSMCs are contractile. However, in response to changing chemical and mechanical signals they change their phenotype, leading to a deregulation of cellular functions. Phenotypic switching is regulated through crosstalk from endothelial cells (EC) to the VSMCs as well as through direct mechanical forces (hypertensive pressure, flow and stiffness of the cellular microenvironment) acting on VSMCs. Importantly, our recent data also demonstrates signalling from the VSMC (once phenotypically switched) to ECs that lead to a change in the barrier function, however details of this regulation are currently unclear. Based on pilot data and literature, we hypothesise that this crosstalk is co-regulated through transfer of mRNAs and miRNAs through exosomes.
This project will set up an artery-on-a-chip model to study this crosstalk in detail. It will be split into three aims:
- Setup of Artery-on-a-chip model to investigate EC-VSMC crosstalk
- Multianalyte analysis of exosomal content, depending on changing mechanical stimuli (i.e. pressure and flow), and effect of respective exosome transfer on EC and VSMC phenotype
- Multianalyte analysis of exosomal content, depending on chemical stimuli (TGF-beta, or known pro-atherosclerotic dietary or other chemicals, such as PCBs and Nicotine), and effect of respective exosome transfer on EC and VSMC phenotype
The outcome of the project will be a new predictive model for arterial disease with improved consideration of the complexity of EC-VSMC interactions.
Developing adrenal chip models of disease for the testing of pharmacological and advanced therapies
Supervisors: Prof Li Chan, Prof John Connelly
Industry Partner: OMass Therapeutics (TBC)
Glucocorticoids (GC), produced by the adrenal glands, are essential for life and necessary to respond to environmental stress, illness, and changes in metabolism. Too little GC can be lethal and too much GC leads to life-long morbidities. GC production is controlled by Adrenocorticotropic hormone (ACTH) secreted from the pituitary gland. ACTH acts on the melanocortin 2 Receptor (MC2R) [also known as the ACTH receptor] along with its accessory protein, the Melanocortin 2 Receptor Accessory Protein (MRAP). The MC2R/MRAP complex are critical for GC synthesis within the adrenal gland. Loss-of-function mutations in the MC2R and MRAP leads to familial glucocorticoid deficiency type-1 and 2 respectively (OMIM 202200, 607398). In contrast, excess GC results in Cushing’s syndrome. As a laboratory we have been developing new therapeutics for the treatment of diseases that affect the adrenal gland, but there are currently limited experimental tools to study human adrenal function and disease mechanisms. The overarching aim of the PhD will be to develop human adrenal models on a chip to enable testing of pharmacological and advanced therapies GC diseases. The student will have the chance to work with one of the most exciting UK biotech companies in this area.
The aims of the PhD are:
- Develop a microfluidic adrenal gland on chip model that can produce GC in response to ACTH.
- Develop an adrenal gland on chip model of relevant diseases resulting in GC excess and GC deficiency.
- Testing and validation of pharmacological agents and novel therapies utilising these models.
Anticipated outcomes:
- Development of new models that will benefit our laboratory, the research community, our industrial partners and ultimately patients that we serve in our clinics.
- Training of a PhD student in the cutting edge of engineering/health biomedical research and industry preclinical space of developing new therapies for health.
A multi-organ model for breast cancer metastasis to bone and liver
Supervisors: Prof Wen Wang, Prof Martin Knight, Prof Stefaan Verbruggen
Industry Partner: CN-Bio
This exciting PhD studentship project will develop new organ-chip models to explore breast cancer metastases to liver and bone. The student will start by developing a primary human breast tumour model and then linking this to a liver model to examine liver metastasis. The student will then develop a bone model and combine this with the primary tumour model to examine bone metastasis. This project builds on existing research from Prof Knight and Dr Verbruggen recently published in Advanced Science and Cancers and our on going research developing and using bone metastasis organ-chip models.
The project is supported by our partnership with the organ-chip company, CN-Bio, and will use their commercial state-of-the-art multi-organ platform available within the Centre. This studentship also includes the option of a placement in CN-Bio's laboratories in Cambridge which will support the research project and provide the student with additional transferrable skills and industrial perspective. The student will be part of a large multi-disciplinary organ-chip research group and will be supported by 3 post docs working on a new EPSRC grant in this area of organ-chip technology (EPSRC grant: Organ-chips for accelerated deployment of new medicines), as well as ongoing collaborations with Barts Cancer Institute.
Development of a human hepatocyte culture for measuring drug metabolism and elimination to replace animal models used in drug testing
Supervisors: Prof Kenny Linton, Dr Adrian Biddle
Industry Partner: TBC
We have developed a Matrigel-sandwich culture to differentiate mature hepatocytes (iHEPs) from iPSCs. The iHEPs form tissue similar to hepatic plates with enclosed canaliculi between adjacent cells (easily visualised using fluorescent CDF transported by MRP2). The iHEPs secrete bile into the canaliculi which is recovered for mass spectrometry following release of the tight junctions.
Our first aim will replace Matrigel with a synthetic extracellular matrix (ECM). The Space of Disse, which surrounds hepatocytic plates, contains ~150 ECM proteins (Arriazu 2014. However, it simplifies significantly around the most mature hepatocytes where it is enriched in fibrillar collagens, fibronectin and sulphated chondroitin and dermatan. Peptimatrix hydrogels ‘RGD’ and ‘Plus’ (mimetics of fibronectin and collagens, respectively), offer an excellent starting material. This will significantly improve the relevance of this organ-in-a-dish model for mimicking liver tissue.
Our second aim will identify the transporter of the hepatocellular cancer (HCC) drug sorafenib (SHARP trial; Llovet 2008 NEJM. HCC is a common malignancy (Ferlay 2018 with a high relapse rate. Sorafenib, a multi-kinase inhibitor targeting both tumour cell proliferation and vascularisation, is eliminated via the liver, implicating the drug efflux pumps of the canalicular membrane. Resistance to sorafenib correlates with MRP2 expression and in patient samples (Neis 2001), however the physiological relevance is unproven. CRISPR-Cas9 will generate a knock-out cell line lacking MRP2 (and loss of CDF accumulation the canaliculi). Measurement of sorafenib in the bile will define the importance of MRP2.
Biosensor functionalised organ-chip platforms for improved versatility of predictive in vitro models
Supervisors: Dr Christopher Chapman, Dr Marco Pensalfini
Industry Partner: TBC
Due to the wide variety of cell culture models used predictive in vitro models and organ-on-a-chip systems, there is often insufficient customisability in commercially available chips. This typically overcome by individual researchers producing their own highly customised platforms, however, this can lead to difficulties in reproducibility across the field. This project aims to overcome this significant hinderance to the standardisation of on-chip platforms, by developing a fully customisable three-dimensional printing method to rapidly produce organ-chip platforms to any desired specification. Additionally, to impart improved data output from the organ-chip platforms, this platform will be developed with the ability to add electrodes in any configuration in the channels.
The project will develop novel three-dimensional printing methods for producing these on-chip platforms. Key research challenges will be enabling high resolution prints with tailored geometries through development of a bespoke printing set-up and post processing protocol. Dr Chapman’s lab is focused on utilising conducting polymers for various biosensor applications, and therefore another key research goal will be to enable electrode patterning during the printing of the organ-chip system. This project will be conducted in close collaboration with Dr Achala de Mel, CEO of NuTissu, who has developed multiple 3D printing inks with tuneable physical and electrochemical properties.
Ultimately the system developed through this project will facilitate improved organ-chip technologies to be used across the entire sector, improve reproducibility, and reduce cost of performing research on bespoke organ-chip systems.
Metabolic and immune interactions in the oral mucosal host-microbial interface via advanced biofilm-organotypic models
Supervisors: Dr Abish Stephen, Simon McArthur, Robert Allaker,
Industry Partner: Haleon plc
The oral cavity’s complex microbiome exists in equilibrium with host tissues but shifts in this balance can lead to diseases such as gingivitis and periodontitis. Volatile organic compounds (VOCs) produced by oral bacteria are increasingly implicated in these processes, yet the mechanisms linking microbial metabolism, microenvironmental gradients, and host responses remain unclear.
This project will develop a flow-based in vitro model that integrates organotypic oral tissue constructs with polymicrobial biofilms. By employing increasingly complex microfluidic platforms, we will simulate physiologically relevant fluid flow over layered mucosal and gingival tissues, maintaining stable gradients and nutrient delivery. To gain real-time insights into the chemical and physical parameters that shape host-microbe interactions, we will incorporate sensor foil-based imaging systems for non-invasive detection of oxygen and pH gradients. Mapping these gradients will reveal how subtle environmental shifts influence bacterial VOC production and host tissue responses. Parallel analyses using gas chromatography-mass spectrometry will identify the specific VOCs produced. Moving beyond commercially available technologies, this model will incorporate immune cell transmigration dynamics into the system, elucidating how these VOCs modulate inflammation and barrier integrity.
We envisage this organ-on-a-chip platform will provide a powerful tool for investigating oral health and disease mechanisms, and as a preclinical screening tool for oral care. By combining tissue engineering, fluid dynamics, and integrated sensing technologies, we aim to uncover new strategies for controlling microbial-driven pathology in the oral cavity.