Tag

Tagged: clinical trials

Sponsored
  • Phase-0 goes mainstream: Evolving from niche concept to core development strategy
  • Economic upside: Reduces attrition and curbs wasted R&D investment
  • Regulatory advantage: Enables earlier, more effective dialogue with global agencies
  • Ethical progress: Safeguards patients while speeding access to new therapies
  • Strategic turning point: Phase 0 positioned to become an industry standard

Phase-0 Goes Mainstream

Drug development is one of the most capital-intensive, high-risk endeavours in modern industry. The cost of advancing a single therapeutic candidate from discovery to market is >$2B, with timelines stretching over a decade. Compounding this burden is an industry-wide attrition rate of ~90%, leaving companies and investors with escalating sunk costs and diminishing returns. The conventional Phase I-IV clinical trial pathway - while responsible for many medical breakthroughs - is showing structural limits in an era that prizes both scientific agility and financial discipline, especially as the once-understated Phase IV stage gains prominence with regulators’ growing demand for real-world evidence.

Amid these pressures, a once-unconventional approach is emerging as a strategic lever: Phase-0 microdosing clinical trials. First codified by the FDA in 2006 under its exploratory Investigational New Drug (IND) framework, Phase-0 was long regarded as a niche tactic with limited application. This perception has shifted. Driven by advances in bioanalytical sensitivity, improved modelling platforms, and growing regulatory endorsement, Phase-0 is now being adopted as a mainstream risk-management tool in early development.

By generating early human data on how a compound behaves and acts, Phase-0 enables sharper portfolio triage, earlier go/no-go decisions, and greater capital efficiency. For investors, this is more than incremental progress - it marks a step-change in how biotech and pharma deploy R&D capital, de-risk pipelines, and accelerate development. What began as a regulatory pilot has become a competitive imperative.

 
In this Commentary

This Commentary explores the rise of Phase-0 clinical trials from a niche concept to a transformative force in drug development. It examines how Phase-0 addresses the twin challenges of cost and attrition, while strengthening ethics, regulatory engagement, and patient advocacy. The thesis is clear: Phase-0 is no longer optional. For investors and innovators, it represents a strategic inflection point - reshaping R&D economics, accelerating timelines, and redefining the path to translational success.
 
The Traditional Clinical Trial Paradigm - The Valley of Death

The traditional clinical trial paradigm - long upheld as the gold standard of drug development - comprises four sequential stages that have remained largely consistent since their formalisation in the mid-20th century. Phase I studies, typically enrolling 20 to 100 healthy volunteers, explore safety, tolerability, and pharmacokinetics: how the body absorbs, distributes, metabolises, and excretes a compound, determining its onset, intensity, and duration of action. Promising candidates then advance to Phase II trials, involving several hundred patients to evaluate preliminary efficacy, refine dosing regimens, and identify side-effect profiles. Phase III represents the pivotal test: large, often multinational trials enrolling thousands of participants to generate the robust, confirmatory data required for regulatory approval. Upon successful completion, a drug may enter the market - but the process does not end there. Phase IV, or post-marketing surveillance, continues to monitor safety and effectiveness under real-world conditions. Given that pivotal trials often draw from relatively narrow and demographically limited populations, regulators are increasingly mandating post-approval studies and real-world evidence to capture long-term outcomes and assess performance across broader, more diverse patient groups.

This phased architecture emerged in an era dominated by small-molecule drugs, when the prevailing regulatory ethos placed a premium on safety, caution, and rigorous linear testing. For its time, the model was appropriate, creating a framework that protected patients and ensured reproducibility. Yet in today’s therapeutic landscape - characterised by biologics, gene therapies, personalised medicine, and digital biomarkers - this model shows its age.

Attrition rates are extremely high, with roughly nine out of ten drug candidates failing somewhere along the clinical pathway, often late in Phase II or Phase III when the sunk costs have climbed into the hundreds of millions. The time pressure is equally challenging: the median journey from first-in-human dosing to regulatory approval exceeds ten years, too long in a world where patients and clinicians want timely innovation. Compounding this is a scientific mismatch - animal models, the bedrock of preclinical validation, are unreliable surrogates for human biology, especially in fields such as oncology, central nervous system disorders, and immunology.

These inefficiencies carry ethical implications. Patients enrolling in early-phase trials often do so with hope, but in reality most will be exposed to experimental compounds that never reach the clinic. The tension between scientific necessity and patient welfare underscores the fragility of the current system.

The result is what has become known as the valley of death in translational medicine - the chasm between discovery and delivery, where promising ideas falter not for lack of ingenuity, but because the system exacts a heavy toll in time, money, and human cost. Bridging this valley has become one of the challenges of modern biomedical innovation. Industry, regulators, and patients are seeking alternatives: new trial designs, adaptive methodologies, real-world evidence, and more predictive preclinical models. The future of medicine may well depend on how effectively we reimagine the pathway that leads from laboratory insight to life-changing therapy.

 
Phase-0 Trials: A First Look at Human Biology

Phase-0 trials - sometimes called exploratory IND studies or microdosing trials - mark a departure from the traditional clinical trial continuum. Conceived to de-risk drug development early, these studies move investigational compounds into humans sooner, but under carefully constrained conditions. Unlike conventional trials that push toward therapeutic dosing, Phase-0 is about exploration rather than treatment. Doses are kept extremely small - typically <100 micrograms, or about one-hundredth of the expected pharmacologically active dose - significantly below any level likely to produce clinical benefit or toxicity.

The purpose is not to test whether a new drug works, but to ask a more fundamental question: how does this compound behave in the human body? Phase-0 studies focus on generating pharmacokinetic (PK) and pharmacodynamic (PD) data, probing how a drug is absorbed, distributed, metabolised, and excreted, and whether it reaches and engages its intended biological target. With small cohorts - often 10 to 15 participants, frequently healthy volunteers - and short durations, these trials provide a first look at human biology in relation to specific compounds.

The doses administered in Phase-0 studies are so small that they pose virtually no safety risk. Yet, this also means conventional clinical endpoints - such as therapeutic effects - cannot be measured. To compensate, these trials rely on highly sensitive analytical technologies capable of detecting minute quantities of the drug and its metabolites. Techniques such as accelerator mass spectrometry (AMS), liquid chromatography-tandem mass spectrometry (LC-MS/MS), and positron emission tomography (PET) make it possible to measure drug levels, tissue distribution, and target engagement with precision. These tools transform what would otherwise be invisible into actionable data.

The contrast with Phase I trials is striking. Whereas Phase I typically involves 20 to 100 participants and escalating therapeutic doses to establish safety and tolerability, Phase-0 pares the process back to its scientific essentials. The goal is not safety confirmation or dose escalation, but an early signal - an insight into whether the drug behaves as predicted in silico and in animal models. The risks are lower, but so too are the ambitions: no one expects therapeutic efficacy at microdose levels.

The strategic value of this approach lies in efficiency. By offering a early “peek into humans” at a fraction of the cost and risk of full-scale early trials, Phase-0 enables developers to make sharper go/no-go decisions before committing resources to large-scale programmes. Promising compounds can be prioritised with confidence, while those that falter can be abandoned earlier, sparing patients unnecessary exposure and investors wasted capital. In an industry where time is money and attrition high, Phase-0 trials represent a bridge across the valley of uncertainty that lies between preclinical promise and clinical proof.
The surgical MedTech industry is shifting from proprietary devices to a connected, data-driven ecosystem. Software-first design, AI, and interoperability are redefining the perioperative journey. The latest episode of HealthPadTalks, From Devices to Platforms, unpacks ten forces driving that change - and why the question isn’t which device you build, but which network you enable.
Why Phase-0 is Becoming Mainstream

For years after the FDA introduced its exploratory IDN guidance in 2006, Phase-0 trials remained a niche tool. That is no longer the case. A convergence of scientific, regulatory, economic, and ethical forces is now propelling Phase-0 into the mainstream as a component of modern drug development.

Technological Breakthroughs Have Removed Previous Barriers
  • Unprecedented sensitivity: Ultra-sensitive methods like Accelerator Mass Spectrometry (AMS) can now detect drug levels at attomolar concentrations. This means researchers can generate pharmacokinetic (PK) profiles from microdoses a fraction of traditional clinical trial doses.
  • Real-time insights: Molecular imaging techniques such as PET scanning make it possible to watch a drug binding to its target and track its distribution inside the body.
  • Actionable biomarkers: New biomarker strategies allow early reliable readouts of whether a drug is engaging its intended biological target - something investors and regulators increasingly demand before capital commitments.
Together, these advances mean Phase-0 results are no longer “exploratory curiosities”, but robust, decision-shaping data.

Regulators Have Endorsed the Approach
  • FDA leadership: The FDA’s eIND framework lowered toxicology requirements for Phase-0 studies, making them faster and cheaper to initiate.
  • Global adoption: The European Medicines Agency (EMA) and Japan’s PMDA have since introduced aligned frameworks.
  • Global harmonisation: With multiple regulators now on board, it is feasible to run coordinated Phase-0 programmes across major markets, making the approach attractive for global pharma pipelines.
This regulatory shift has de-risked adoption for sponsors and provided a playbook for execution.

The Economics Are Compelling
  • Cost avoidance: The average cost of advancing a drug to Phase II can reach hundreds of millions of dollars. If Phase-0 data reveal poor pharmacology early, companies can exit that programme for only a few million.
  • Capital efficiency: The Phase-0 model frees resources to be redeployed into higher-probability candidates, shortening timelines and improving ROI.
Phase-0 offers one of the best early filters for drug development risk - something every R&D-intensive business needs.

A Patient-First Model Aligns with Ethics and Market Demands
  • Minimal exposure, maximum learning: Patients are exposed to microdoses significantly below therapeutic levels, dramatically lowering risk.
  • Transparency and trust: Patient advocacy groups are pushing for faster, more efficient trials. Phase-0 resonates because it avoids wasting patient participation on drugs that were never likely to succeed.
This alignment with ethical imperatives makes Phase-0 attractive not just to regulators, but to patients, advocacy groups, and public opinion.

Perfect Fit for Modern Drug Pipelines
  • Precision oncology: Complex, personalised cancer drugs need early human validation of mechanism. Phase-0 provides this.
  • CNS therapies: Brain drugs face unique delivery and engagement challenges; Phase-0 with imaging can confirm penetration and binding.
  • Biologics and novel modalities: As pipelines diversify into antibodies, RNA therapeutics, and beyond, Phase-0 becomes a tool to validate mechanism without high-risk investment.
Phase-0 aligns well with the needs of today’s most valuable drug classes.

Phase-0 is no longer experimental - it is becoming standard practice. It combines technological readiness, regulatory acceptance, economic necessity, patient alignment, and therapeutic relevance into one package. The companies that adopt Phase-0 early gain a competitive edge: they can kill failures faster, invest more confidently in winners, and deliver innovative therapies to patients with greater efficiency.

 
Case Studies: Phase-0 in Action

Oncology Cancer drug development has been an early adopter of Phase-0 methodologies. For instance, PET microdosing has been applied to assess tumour penetration of kinase inhibitors prior to therapeutic escalation. Such approaches allow researchers to prioritise compounds with the most favourable tissue exposure profiles, reducing the risk of late-stage attrition.

Neuroscience In central nervous system (CNS) drug discovery, the blood–brain barrier (BBB) remains a challenge. Phase-0 studies integrating microdosing with PET tracers have provided early evidence of whether candidate antidepressants and antiepileptics achieve adequate brain penetration. This enables developers to discontinue non-viable molecules earlier, conserving resources and avoiding unnecessary patient exposure.

First-in-class agents  Novartis has underscored the strategic and financial value of Phase-0 studies in optimising R&D efficiency. By integrating exploratory microdosing into its early development process, the company was able to rapidly identify the most promising kinase inhibitor candidates. This data-driven approach not only accelerated pipeline decisions but also reportedly saved multiple years of development time and millions in downstream investment.

Academic consortia The Microdosing Network has spearheaded collaborative Phase-0 initiatives across academic medical centres. These efforts have not only broadened access to the methodology but also fostered greater transparency and public trust in early-stage drug research.

Across oncology, neuroscience, first-in-class innovation, and academic collaborations, Phase-0 has proven to be a practical, evidence-based component of contemporary drug development pipelines.

 
Benefits of Mainstream Phase-0

1. Scientific Advantages Phase-0 studies generate human pharmacokinetic and pharmacodynamic (PK/PD) data before traditional Phase I. This strengthens translational accuracy by:
  • Demonstrating early how a compound behaves in the human body.
  • Clarifying dose-exposure relationships and confirming whether the drug reaches its intended tissue targets.
  • Significantly reducing the risk of advancing a drug candidate with flawed assumptions.
2. Regulatory Advantages By engaging regulators with concrete human data upfront, companies can:
  • Open a more collaborative, constructive dialogue at the earliest stage.
  • Design more adaptive trials, as Phase-0 findings often inform and refine Phase I protocols.
  • Potentially accelerate regulatory feedback cycles, streamlining approvals downstream.
3. Financial Advantages For investors, Phase-0 offers an economic filter:
  • Candidates with little chance of success are identified within months, not years, preventing the waste of hundreds of millions.
  • Eliminates premature investment in large-scale synthesis, toxicology, and manufacturing infrastructure for drugs unlikely to succeed.
  • Enables portfolio optimisation, reallocating resources toward winners earlier and with greater confidence.
 4. Ethical Advantages Ethics align with economics:
  • Patients are shielded from exposure to compounds that early human data suggest are ineffective or unsafe.
  • Transparency and prioritisation of safety build greater trust among patients, advocacy groups, and the public - strengthening the reputation of sponsors and investors.
5. Operational Advantages From a business execution perspective, Phase-0 is transformative:
  • Critical go/no-go decisions can be made in months instead of years.
  • Multiple drug candidates can be tested in parallel at minimal cost, allowing companies to pursue a "shots-on-goal" strategy without diluting resources.
  • Development timelines are streamlined, improving capital efficiency across the R&D pipeline.
6. Patient and Advocacy Alignment The patient voice in drug development is becoming louder. Advocacy groups demand faster, more efficient progress toward effective therapies. Phase-0 is responsive to this pressure:
  • By filtering out “dead-end” drugs earlier, timelines to efficacious treatments are shortened.
  • This positions companies as responsive, responsible partners in the shared mission of accelerating cures - an important differentiator in the eyes of patients, payers, and policymakers.
HealthPadTalks is a podcast exploring the trends redefining healthcare’s future. Building on HealthPad’s Commentaries, we don’t just deliver answers — we question them. Through bold ideas, diverse voices, and meaningful debate, we aim to improve outcomes, cut costs, and expand access for all. Make sure to follow us! 
Challenges and Limitations

While Phase-0 offers advantages, it is not a universal solution. Its value lies in strategic deployment, and investors should understand both its boundaries and its growing potential.

Scientifically, Phase-0 studies have limitations. Microdose pharmacokinetics (PK) may not always scale to therapeutic doses - particularly in drugs with nonlinear kinetics or saturable metabolism. Similarly, large biologics often do not behave proportionally at sub-therapeutic exposures, meaning Phase-0 may have less relevance in those categories. These are caveats that highlight the need for smart candidate selection rather than undermining the model itself.

On the regulatory front, global alignment is still in progress. While the FDA, EMA, and Japan’s PMDA all endorse Phase-0 approaches, harmonisation across jurisdictions is incomplete, and smaller regulatory agencies often lag. This fragmentation can complicate multinational development strategies, though early adopters who navigate it effectively gain a competitive edge.

Operationally, the specialised tools required - such as accelerator mass spectrometry (AMS) and advanced PET imaging - come with costs and infrastructure demands. Recruitment also presents challenges, since participants in Phase-0 studies do not receive direct therapeutic benefit. That said, as the ecosystem matures, central labs and contract research organisations (CROs) are expanding access to these capabilities, lowering barriers to entry over time.

Ethically, some scholars raise concerns about exposing volunteers to compounds with no therapeutic intent, even at very low doses, suggesting tension with traditional consent frameworks. Yet regulatory agencies and ethics committees increasingly accept the practice when safety is rigorously managed, especially as patients and advocacy groups push for faster, safer drug development pathways.

Finally, cultural resistance within parts of the pharmaceutical industry persists. Established organisations can favour “tried and tested” approaches, viewing Phase-0 as unnecessary. This conservatism is eroding as case studies demonstrate that early human data can prevent multi-hundred-million-dollar failures. For investors, this cultural inertia is both a headwind and an opportunity: companies that adopt Phase-0 ahead of the curve can create a competitive advantage.

 
The Future Outlook: Phase-0 in the Next Decade

Over the coming decade, Phase-0 trials are set to move from a niche strategy to a mainstream pillar of drug development. For investors, this represents a scientific transformation and a structural shift in how capital is deployed, risks are managed, and timelines compressed.

One of the most significant trends will be the integration of Phase-0 into adaptive trial designs. Instead of being a standalone experiment, microdosing studies will increasingly serve as essential steps to Phase I, creating a continuous data flow that accelerates progression while reducing uncertainty. Such integration means capital is no longer “parked” for years before meaningful inflection points; it is working harder and delivering answers faster.

AI will amplify these advantages. By applying predictive models to Phase-0 data, companies will sharpen candidate selection and identify winners earlier. The combination of human microdose data with AI-driven analytics could transform the probability of success across pipelines, making Phase-0 not just a filter but a proactive optimisation engine.

Personalised medicine will also benefit. Microdosing studies provide a safe, low-risk way to stratify patients based on pharmacogenomics or biomarker profiles. This could enable drug developers to understand who a therapy works best for before scaling investment - aligning with precision medicine trends and payer demands for demonstrable value.

In rare diseases, where every patient is precious and recruitment a bottleneck, Phase-0 can optimise scarce resources. By clarifying early which compounds warrant full development, developers avoid wasting limited patient cohorts on drugs unlikely to succeed, thereby preserving opportunities for promising therapies.

Regulatory convergence is another catalyst. By 2035, we can expect much greater harmonisation across major agencies making Phase-0 a globally consistent tool. Companies that position themselves now will be well placed to capitalise on this alignment, gaining smoother multinational pathways.

Perhaps most importantly, Phase-0 is already showing strength in oncology, central nervous system disorders, and advanced biologics. In these areas, where development costs are steep and patient need is urgent, Phase-0 is likely to become as routine a starting point as Phase I initiation.

For investors, the trajectory is clear: Phase-0 is evolving from an experimental option into a core component of the drug development ecosystem. Those who recognise and back this shift early will benefit from improved R&D economics, and from the reputational upside of enabling faster, safer, and more precise therapies for patients worldwide.

 
Takeaways

Phase-0 clinical trials, once regarded as experimental, are now redefining the architecture of drug development. They confront the twin crises undermining pharmaceutical R&D - escalating costs and high attrition - while aligning with a growing ethical imperative: to protect patients and hasten the delivery of effective therapies. For investors and innovators, this shift transcends incremental efficiency; it signals a transformation in the economics of innovation.

As the scientific, regulatory, and cultural ecosystems mature, Phase-0 is poised to evolve from a tactical advantage into a foundational norm. The next generation of competitive pipelines will embed Phase-0 not as an option, but as a prerequisite - reducing waste, de-risking capital, and compressing timelines. As this paradigm becomes integral to the early stages of development, the cumulative effect will be substantial: the cost of bringing new drugs to market will fall, enabling more affordable access to life-changing treatments for millions of patients.

For the pharmaceutical industry, this represents a moment of strategic inflection. By championing and operationalising Phase-0, companies can position themselves not merely as participants in drug development, but as architects of a more equitable healthcare future - one where efficacy, safety, and accessibility are not competing priorities but shared outcomes. Start-ups, too, have a unique opening: by coupling Phase-0 insights with advances in AI and machine learning, they can become indispensable accelerators of translational discovery.

Ultimately, the future of clinical research may no longer begin with a costly leap into Phase I, but with a measured, data-rich step into Phase-0 - a step that promises smarter science, safer patients, and a fairer world. In this evolution lies the possibility that access to efficacious treatments - and the closure they bring - becomes not a privilege of circumstance, but a universal human right.
view in full page
joined 11 years, 7 months ago

Jalid Sehouli

Professor and Director, Department of Gynecology Campus Virchow Klinik and Campus Benjamin Franklin, Charité Medical University Berlin; Vice-President, EUTROC network

Professor Sehouli is a gynaecological oncologist, working at the Charité, University of Medicine in Berlin, Germany.


Professor Jalid Sehouli is currently the director of the department of gynecology in the famous Charité/ Campus Virchow-Klinikum and Campus Benjamin Franklin. He is also the head of the European Competence Center for Ovarian Cancer and is leading the gynaecological oncology at all three Campi at Charité Medical University in Berlin.


Professor Sehouli founded the German Foundation for ovarian cancer patients, being involved in many initiatives for patients.

Together with Professor Lichtenegger he founded the German north eastern society for gynaecological oncology (NOGGO). In 2000 he initiated the tumor bank ovarian cancer, which became the largest biobank for fresh frozen tissue worldwide.

Since 2013 Professor Sehouli was nominated as speaker for the OVAR Kommission of the AGO, Germany.


His clinical and scientific interest focuses on the surgical therapy and systemic treatment of advanced gynaecological malignancies. He lead several preclinical trials in predicting the surgical outcome. He is principle investigator in many Phase I to Phase III clinical trials in ovarian cancer.


He has published more than 250 national and international papers in the field of gynaecologic oncology.
He is a member of the executive board of the Arbeitsgemeinschaft Gynäkologische Onkologie (AGO) and the Nord-Ostdeutsche Gesellschaft für Gynäkologische Onkologie (NOGGO), and a member of the European Society of Gynaecological Oncology (ESGO), the Gynaecologic Cancer Intergroup (GCIG), the American Society of Clinical Oncology (ASCO) and the Multinational Association of Supportive Care in Cancer (MASCC).


view this profile
joined 11 years, 7 months ago

EUTROC

European Translational Research network in Ovarian Cancer

EUTROC (European Translational Research network in Ovarian Cancer) is a European cancer network addressing the fragmentation currently hindering translational research for cutting edge therapies in ovarian cancer.

It will strengthen European excellence in the diagnosis and treatment of OC.

The need to bring promising drug candidates and treatments into clinical settings is currently delayed by the lack of standardised protocols for preclinical animal studies, lack of suitable ovarian cancer models, lack of validated predictive biomarker diagnostics, and integrated appropriate management of translational research partners (including pharma and biotech companies).

Developing effective diagnostic and treatment strategies require structured joint efforts of experts on tumor immunology, epigenetics, pathology, bioinformatics, drug development and delivery, trial development, clinical oncology and gynecological oncology.

Leading European researchers and clinicians together with the pharmaceutical industry and biotech companies will focus on preclinical and clinical studies in ovarian cancer. Therefore new targets will be identified, validated and translated to new drugs and brought to clinical practice. Standardisation and harmonisation of laboratory practices and good clinical practice procedures will BE performed within the consortium.

The consortium combines expertise in clinical as well as preclinical research. Involved researchers are coordinating a number of investigator-initiated Phase I, II trials.The Network will build a platform for research and strong educational programs for the next generation of experts aimed at developing individualised patients benefit.


view this profile
joined 12 years, 5 months ago

Mike Birrer

Vice chancellor and director, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences

Dr Birrer is vice chancellor and director of the Winthrop P. Rockefeller Cancer Institute at the University of Arkansas for Medical Sciences (UAMS).

Birrer completed his medical degree and doctorate of philosophy in 1982 in the Medical Scientist Training Program at the Albert Einstein College of Medicine in New York. Following a medical internship and residency at Massachusetts General Hospital, Birrer entered the Medical Oncology Fellowship program at the National Cancer Institute in Bethesda, Maryland. After his fellowship, Birrer was appointed senior investigator (with tenure) and established the molecular mechanism section in the Division of Cancer Prevention and Control.

In 2008, Birrer was appointed professor of medicine at the Harvard School of Medicine and assumed the position of director for both Gynecologic Medical Oncology at Massachusetts General Hospital and the Gynecologic Oncology Research Program at the Dana Farber/Harvard Cancer Center.

In 2017, he accepted the position of director of the O’Neal Comprehensive Cancer Center at the University of Alabama at Birmingham, where he served as professor of medicine, pathology and OB-GYN.

Recognized nationally and internationally as an expert in gynecologic oncology, Birrer’s primary research interest is in characterizing the genomics of gynecologic cancers to improve the clinical management of these diseases. His clinical interests include ovarian cancer, endometrial cancer and cervical cancer.

Birrer has approximately 400 publications, including peer-reviewed manuscripts, book chapters and review articles. He served as chair and chair emeritus of the Department of Defense Ovarian Cancer Research Program, chair of the Committee for Experimental Medicine of the Gynecologic Oncology Group, chair of the Translational Science Working Group of the Gynecologic Cancer Intergroup, and a member of the Gynecologic Cancer Steering Committee.


view this profile