Dr. Aditi Mehta, Director and Head of Innovation at
Merck KGaA – mRNA and LNPs
Dr. Aditi Mehta, Director and Head of Innovation – mRNA and LNPs
Biography
Aditi leads the Innovation Department focusing on mRNA and LNP technologies within the CDMO Business of Merck KGaA, Darmstadt, Germany. Her team is dedicated to advancing mRNA design and developing scalable and flexible manufacturing processes, developing novel excipients for efficient and safe mRNA delivery, and optimizing LNP manufacturing processes, with a strong emphasis on product quality and sustainability. Aditi is a Biologist by training and earned her MSc in Genetics and her PhD in Molecular biology. Subsequently, Aditi gained valuable postdoctoral experience in nucleic acid delivery under the mentorship of Prof. Dr. Olivia Merkel at the Department of Pharmacy, LMU Munich. Following her postdoctoral work, she spent two years as a Group Leader in the Team of Prof. Dr. Merkel and was awarded the LMU Early Career Grant ("Der Nachwuchsförderungsfonds") and the Joachim Herz Stiftung Fellowship for Interdisciplinary Research. Aditi has over 10 years of experience in the interdisciplinary field of nanomedicine and RNA delivery, working with both polymeric and lipid-based delivery vehicles. Her research primarily focused on lung cancer and the targeted delivery of nucleic acids during her time at LMU.
Interview
NanoSphere: Tell us a bit about yourself—your background, journey, and what led you to where you are today.
Aditi: My career trajectory, while seemingly shaped by a series of fortuitous events, has been deeply influenced by a growing passion for genetics and gene therapy. I grew up in New Delhi, India, with aspirations of becoming a doctor, like most of my family. However, during high school biology, I was introduced to the field of genetics, which immediately captured my interest. This enthusiasm was further ignited during my undergraduate studies, particularly by a guest lecture on human genetics by Prof. BK Thelma, sparking my desire to delve deeper into the field. I pursued a Master of Science in Genetics at the University of Delhi, followed by a PhD in Molecular Biology, specializing in lung cancer, at the Max Planck Institute for Heart and Lung Research. Toward the end of my PhD, I conducted experiments on siRNA delivery to the lungs using polyethyleneimine (PEI) as a non-viral delivery vehicle, which led me to appreciate RNA's potential to target previously "undruggable" diseases/targets. However, at the time, I had limited understanding of how to deliver mRNA or siRNA therapeutics beyond my initial experience with one type of delivery vehicle. A turning point in my career came when I met Prof. Dr. Olivia Merkel at a conference, who went on to become a major influence and mentor in my professional journey. I was fortunate to join her team as a postdoctoral researcher at the Department of Pharmacy, LMU Munich, where I focused on nucleic acid delivery to the lung. Although transitioning to this new field after completing my PhD felt like a risk, it was a decision I’ve never regretted. During my postdoc, I immersed myself in learning everything I could about RNA delivery technologies, fascinated by the immense promise and transformative potential of mRNA therapeutics. In early 2020, I began seeking new career opportunities, and, quite unexpectedly, amidst the height of the COVID-19 pandemic, I discovered an ideal position— Senior manager, RNA Delivery at Merck KGaA, Darmstadt, Germany. I joined Merck in September 2020, and over the past few years, my role has evolved to encompass the Innovation and R&D functions related to mRNA manufacturing, excipient design and synthesis, mRNA delivery, and LNP manufacturing. This role perfectly integrates my foundational education in genetics and molecular biology with my postdoctoral expertise in RNA delivery, enabling me to contribute to cutting-edge advancements in the field.
NanoSphere: Given your experience leading innovation in mRNA and LNP technologies, what broader scientific or technological trends do you find most promising for improving the next generation of RNA delivery—especially in terms of scalability, stability, or targeting?
Aditi: The next generation of RNA delivery technologies holds immense promise, especially as the field continues to evolve. One of the most promising areas is the optimization of lipid nanoparticles (LNPs), particularly ionizable lipids, which are critical for the efficient and stable delivery of mRNA. The design of next-generation LNPs that offer enhanced stability, improved biocompatibility, and reduced immunogenicity is a major focus. Innovations in lipid composition could lead to more efficient mRNA delivery with lower doses. As LNP formulations are refined, the challenge remains to achieve better tissue specificity and reduce off-target effects, which could be addressed through further functionalization and targeted delivery strategies. In addition, looking into non-lipid nanocarriers (such as polymeric nanoparticles or peptide-based delivery) as alternatives or complementary approaches to LNPs could offer greater tissue specificity, reduce systemic toxicity, and improve the control of drug release. One of the persistent challenges with RNA therapeutics is mRNA stability and degradation. Advances in chemical modifications to the RNA structure, such as novel cap structures, nucleotide modifications or incorporating modified bases, can help stabilize mRNA, reduce immune activation, and improve translation efficiency. Further, advancements in self-amplifying RNA or circular RNA may allow for more sustained expression of the encoded proteins, making them more effective for chronic or prolonged treatments. The role of AI and machine learning is also growing in RNA therapeutics, enabling more efficient design of RNA molecules, selection of optimal delivery vehicles, and formulation optimization. By leveraging high-throughput screening and data analytics, AI can help predict the best mRNA sequence and formulations for specific therapeutic applications, accelerating the discovery of novel RNA therapeutics. On the manufacturing side, scalable and modular RNA production platforms are becoming increasingly important. Innovations in cell free DNA template generation, in vitro transcription (IVT) and continuous manufacturing systems are making it possible to produce high quality mRNA at larger scales, ensuring that RNA-based therapies are cost-effective and more accessible.
NanoSphere: Given the rapid growth of the field and the increasing demand for expertise, how quickly do you think a fresh PhD—can ramp up and begin contributing independently?
Aditi: A well-trained PhD can typically begin contributing independently within 6-12 months after graduating, depending on how closely the new role aligns with their existing expertise. If the new challenge requires additional competencies or skills, the timeline for becoming fully independent may extend, but with the right training and mentorship, progress can be rapid. In my (admittedly biased) opinion, stepping outside one’s comfort zone and venturing beyond their area of expertise often opens up invaluable development opportunities. While it may feel uncertain and time consuming at first, the opportunities for growth, skill development, and broader perspective often outweigh the challenges, leading to long-term success and enhanced capabilities.
Aditi: A well-trained PhD can typically begin contributing independently within 6-12 months after graduating, depending on how closely the new role aligns with their existing expertise. If the new challenge requires additional competencies or skills, the timeline for becoming fully independent may extend, but with the right training and mentorship, progress can be rapid. In my (admittedly biased) opinion, stepping outside one’s comfort zone and venturing beyond their area of expertise often opens up invaluable development opportunities. While it may feel uncertain and time consuming at first, the opportunities for growth, skill development, and broader perspective often outweigh the challenges, leading to long-term success and enhanced capabilities.
NanoSphere: As someone leading innovation in this space, what emerging areas in LNP design or RNA modification are you most intellectually curious about—even outside the traditional pharma lens?
Aditi: Some of the most inspiring research fields in RNA and LNP are pushing the boundaries of chemistry, synthetic biology, nanotechnology and systems design. These involve deep conceptual challenges, unexplored biology and bio-inspired or bio-integrated engineering. The future of RNA isn’t just about discovering new payloads — it’s about delivering them with precision, consistency, and adaptability across indications. A major shift is happening in manufacturing technologies. We need flexible, platform-based systems that can scale rapidly and support multiple RNA formats — from mRNA and saRNA (self-amplifying RNA) to circular RNA — without extensive retooling. That means designing for speed, reproducibility, and global use from the start. Equally important is rethinking delivery vehicles and excipients. In RNA therapeutics, the delivery system is half the drug — sometimes more. LNPs are not just passive carriers; they determine where the RNA goes, how it’s protected, how it enters cells, and how the immune system responds. In many ways, the lipid system is as critical as the RNA itself. Designing next-generation lipids — using AI, combinatorial chemistry, or bio-inspired systems — is now central to mRNA delivery innovations. I personally find ‘precision mRNA delivery’ particularly inspiring, using programmable LNPs that for instance would respond to pH, temperature, light, enzymes, or specific biomarkers to release their cargo precisely where needed – with little to no side effects, and thereby improving patient quality of life. Beyond the traditional pharma lens, I’m particularly curious about the non-pharmaceutical applications of RNA-LNPs. For example, in synthetic biology, RNA can be used as a tool to engineer biosensors that detect environmental pollutants, pathogens, or even changes in biological systems. Imagine using RNA-LNPs as part of to bio-monitor environmental conditions or food safety real-time. These systems could be more cost-effective and scalable compared to traditional detection methods.
NanoSphere: If there’s one key message or insight you’d like to share with readers about the future of nanomedicine, what would it be?
Aditi: The future of nanomedicine isn’t smaller – it’s smarter. The future lies in designing scalable, programmable platforms – where excipients are not just inactive ingredients, but functional, tunable components that drive delivery, stability and performance. But getting there, needs us scientists, engineers and regulators to work together. Innovation only scales when we collaborate across fields and build the future together!
Aditi: The future of nanomedicine isn’t smaller – it’s smarter. The future lies in designing scalable, programmable platforms – where excipients are not just inactive ingredients, but functional, tunable components that drive delivery, stability and performance. But getting there, needs us scientists, engineers and regulators to work together. Innovation only scales when we collaborate across fields and build the future together!