Biotech Valuation Beyond the Hype. A Guide to Reality-Based Assessment

Biotech Valuation Beyond the Hype. A Guide to Reality-Based Assessment

This analysis builds upon my previous articles on disciplined biotech valuation. The series began by establishing risk-adjusted Net Present Value (rNPV) and Real Options Analysis as critical tools, moving beyond traditional finance for preclinical and early-stage biotech assets. Next, we explored how Real Options Analysis quantifies the strategic value of managerial decisions to defer, expand, or abandon R&D programs as new data emerges. Most recently, we detailed a two-layer valuation architecture for platform versus single-asset companies. In that framework, Layer A captures the value of specific programs, while Layer B monetizes a platform’s inherent operational efficiencies, such as increased probabilities of success and business development optionality.

This article focuses on the commercial cornerstone of Layer A valuations: S0, the projected present value of post-launch cash flows. This figure is frequently presented without a clear audit trail, disconnected from the regulatory, operational, and evidence-based realities that dictate commercial success. My goal is to deconstruct S0 into a transparent "glass box." This involves dissecting projected value into its core components: pricing corridors based on payer frameworks, adoption curves grounded in empirical data, launch sequences dictated by geographic and regulatory timelines, and evidence thresholds that link clinical outcomes to market access.

We will then integrate this transparent S0 into a broader governance framework. This includes respecting baseline clinical transition probabilities (priors) and defining the specific evidence required to improve them (posteriors). We will also explore how to translate clinical trial observations into health system funding and how to overlay business development terms, such as option fees, milestones, and profit-sharing, onto this base valuation without double-counting. The result is a complete, auditable architecture for valuing early-stage biotech assets, grounded in evidence that you can actually use.


1. The Glass Box, Not the Black Box

A biotech valuation is often compromised from the start by a single, opaque number presented as S0, the projected value of post-launch cash flows. This figure, offered without a transparent methodology, creates an illusion of certainty. If one cannot audit the patient population estimates, net price derivations, adoption speed, or patent cliff assumptions, the valuation is fundamentally unsound.

The alternative is the "glass-box" S0. This approach deconstructs commercial value into a series of auditable components, each with defined sources, ranges, and potential for error. It transforms high-level disagreements into specific, evidence-based discussions about key parameters.

At its core, S0 is a product of several explicit variables: eligible patients by geography, net price after all concessions, market penetration over time, therapy duration, and operating margins, all framed by a defined Loss of Exclusivity (LOE) schedule. The glass-box method simply demands that the work behind each variable is shown.

Establishing Realistic Price Corridors

Wishful thinking often resides in price assumptions. It is crucial to model net price, not list price.

  • In the United States, significant rebates are standard. The Congressional Budget Office has documented that rebates for brands in Medicare Part D reached approximately 35% by 2018, with some estimates for the broader market approaching 48% [1]. For physician-administered drugs, reimbursement is based on Average Sales Price (ASP), which already reflects discounts and is typically lower than the initial Wholesale Acquisition Cost (WAC) [2]. A rigorous S0, therefore, uses distinct price corridors for pharmacy and medical benefit channels.
  • In Europe and the UK, similar constraints apply. Germany’s AMNOG process leads to negotiated reimbursement amounts based on demonstrated added benefit [3], while England’s NICE framework uses confidential commercial agreements to align prices with cost-effectiveness thresholds [4]. Managed-entry agreements are now the standard across Europe, ensuring that actual prices paid are consistently below list prices [5]. This principle holds even for advanced therapies, where confidential discounts have been central to market access for treatments like Zolgensma and axi-cel [8, 9].

Article content

Figure 1. Adoption ramp archetypes by indication class and payer environment. Penetration trajectories differ systematically across four classes. Oncology biomarker drugs show constrained Year 1 volumes (e.g., Sotorasib, ~850 eligible patients/year under England‘s Cancer Drugs Fund) with step changes following first‑line expansions. Rare disease launches clear prevalent backlogs rapidly (30–60% of steady state in Year 1 in centralized systems), then converge to incident flows. Chronic prevalent diseases face programmed ramps: NICE templates project 20%, 35%, 50% adoption in Years 1–3; US pharmacy‑benefit classes sit lower until price‑to‑value alignment relaxes prior authorization. Ultra‑rare centralized therapies show steep initial uptake limited by small populations and registry commitments. Archetypes calibrated to NICE resource‑impact assessments, Cancer Drugs Fund data, and US payer utilization management.

The Institute for Clinical and Economic Review (ICER) provides further anchors. Its value-based price benchmark for semaglutide suggested a 44–57% discount from WAC would be required to reduce access friction, a clear signal that must be incorporated into any credible forecast [6].

Modeling Market Erosion and Adoption Ramps

The commercial lifecycle does not last forever. An honest S0 must parameterize both the inevitable patent cliff and the realities of market adoption.

Loss of Exclusivity (LOE): The impact of LOE is well-documented and severe. For small molecules, the FDA reports that prices can fall by over 95% once six or more generic competitors enter the market [11]. Biologics erode differently, with biosimilar uptake varying widely, but the impact remains significant [12]. AbbVie’s 45.3% year-on-year revenue decline for Humira in the first year of U.S. biosimilar competition serves as a stark reminder of these dynamics [13]. A glass-box model does not speculate about erosion; it parameterizes it with sourced data.
Article content

Figure 2. Erosion dynamics following loss of exclusivity. Small molecules exhibit crushing nonlinearity: with six or more generic entrants, prices fall >95% below pre‑LOE benchmarks, concentrated in 12–24 months. Biologics erode more slowly. Medicare Part B biosimilar uptake ranges 26–80% by molecule; payment limits sit 13–70% below reference products. NHS switching policies target 100% of new starts within three months and 80% of legacy patients within ten months. AbbVie‘s Humira declined 45.3% year‑on‑year in the first year of US biosimilar entry. Erosion schedules incorporate legal calendars (Orange Book, BPCIA 12‑year baseline) and realized dynamics (generic entry counts, biosimilar penetration ramps). Data: FDA competition analysis, Medicare Part B.

Adoption is also not a generic curve; it follows distinct patterns based on indication and operations.

  • Rare Diseases: Launches can quickly capture a prevalent patient backlog before settling into a flow of incident cases [8].
  • Chronic Diseases: Uptake is often slow and programmed, governed by payer friction and clinical capacity. NICE resource-impact templates, for instance, have projected adoption at 20%, 35%, and 50% over three years for certain chronic therapies [14].
  • Oncology: Adoption depends on label scope and testing infrastructure. A biomarker-gated drug for a late-line setting will have a much smaller initial pool (e.g., sotorasib in England [17]) than a therapy approved for a first-line population with established diagnostics (e.g., Tagrisso [15, 18]).

Accounting for Geography and Time

A disciplined model cannot treat the world as a single market. Geography is time. FDA approvals in oncology, for example, frequently precede EMA decisions by several hundred days. In the UK, routine availability can lag regulatory marketing authorisation by nearly ten months, driven by HTA and commissioning cycles. These delays have a material impact on present value.

Finally, a glass-box S0 makes every assumption traceable. A vague assertion like "we project 35% uptake in Year 2" becomes an auditable statement: "Mid-case UK adoption is 35% in Year 2, anchored to the NICE resource impact template for this class [14], while the U.S. case is adjusted downward to reflect prior-authorization friction unless net price aligns with ICER’s benchmark corridor [6]."

This approach yields a calibrated, transparent S0. It is an essential foundation for valuation. However, this model does not mitigate early-stage risk. The forces that truly govern preclinical and early clinical value are priors, volatility, and finite development horizons. In the following sections, I will place S0 in its proper context and focus on these dominant factors.


2. The Limits of S0: Priors, Risk, and the Finite Horizon

In a theoretical world of perpetual options with no cost to waiting, an investor would never exercise. This mathematical elegance is a poor guide for drug development, which operates under the unyielding pressures of time, competition, and finite resources. For early-stage programs, even the most meticulously constructed S0 is merely a reference point.

The actual value of assets at the preclinical or early clinical stage is governed by three dominant forces:

  • The probability of success (PoS) that can be earned through superior program design.
  • The discontinuous risk that materializes at key data readouts and regulatory gates.
  • The finite horizon that imposes a real and calculable opportunity cost on any delay.

The Primacy of Priors

An impressive S0 is irrelevant if a program is unlikely to ever reach the market. The starting point for any realistic valuation must be empirical priors, or baseline probabilities of success. Industry-wide data from 2011–2020 provide a sobering foundation: the likelihood of a program advancing from Phase I to approval is only about 8% [20]. This varies by modality and therapeutic area, but serves as a crucial anchor.

Moving a program’s value beyond this prior is not achieved by inflating S0, but by intelligent design. Research shows that integrating validated, prospectively-defined biomarkers can roughly double the overall likelihood of approval, with the most significant impact seen at the Phase II transition [21]. Compounding this challenge are the timelines; median durations for Phases I, II, and III are approximately 1.6, 2.9, and 3.8 years, respectively [21]. An asset valuation that prioritizes S0 over these fundamental probabilities has the relationship backward.

Article content

Figure 3. Clinical transition probabilities and development timelines. Phase I→II: 52%; Phase II→III: 28%; Phase III→approval: 56%; overall Phase I→approval: ~8%. Oncology sits below average; vaccines and certain biologics above. Biomarker‑anchored enrichment can double overall approval likelihood, with largest lift at Phase II. Median durations: Phase I 1.6 years, Phase II 2.9 years, Phase III 3.8 years. Priors define starting beliefs; posteriors from Clinical Evidence Modeling and Bayesian Assurance reflect program‑specific design and evidence quality. Data: BIO/Informa 2011–2020 cohort.

Understanding Discontinuous Risk

Uncertainty in biotech does not manifest as a smooth curve. It behaves like a series of discrete, high-impact events—or "jumps"—at pivotal moments like IND, Phase I/II/III data readouts, regulatory decisions, or HTA reimbursement outcomes. While modeling continuous volatility (σ) is useful, what truly matters for governance is the magnitude and probability of these jumps.

The purpose of methods like Clinical Evidence Modeling and Bayesian Assurance (explored later) is to make this uncertainty governable. They provide a posterior PoS, an updated probability of success based on program-specific evidence that answers the critical investment question: Does the next tranche of capital have a credible chance of clearing the next evidentiary hurdle? S0 offers no answer; only a posterior PoS can.

q: The Explicit Cost of Delay

Drug development programs always lose value by waiting. This cost of deferral, known as the convenience yield (q), is a tangible factor that must be accounted for in any dynamic valuation model. It is composed of three main elements:

  • Patent Headroom (q_patent): Every month of delay is a month of lost exclusivity post-launch. This can be estimated by the remaining years of effective patent life and statutory exclusivities, such as the 12-year baseline for biologics [22, 23, 24, 25].
  • Competition (q_comp): In crowded therapeutic areas like oncology or immunology, delays increase the risk that a competitor will establish the standard of care, shrinking a program's potential market share and pricing power.
  • Payer Tightening (q_payer): Health system requirements become stricter over time. In the U.S., the Inflation Reduction Act introduced a clear timeline for drug price negotiations [26, 27]. In the EU, the move toward Joint Clinical Assessments hardens the evidence required for market access [28]. Delaying a program pushes it closer to these more stringent evaluation regimes.

A Credible Valuation Is Governed by Evidence, Not Forecasts.

S0 does not rescue an early-stage asset. Its value is determined by its posterior PoS and its ability to navigate discrete risk events. The finite horizon, quantified by q, dictates the cost of inaction. Therefore, to increase a program's value, the focus must be on superior design—enrichment strategies that improve PoS, translational science that increases confidence, and operational excellence that protects the development timeline.

At early stages, S0 should be used to establish credible commercial boundaries, not to drive investment decisions. The real bets are placed on posterior PoS, jump risk, and time.


3. Geography Is Time: How Regional Timelines Shape Present Value

In financial models, geography is often treated as a static variable: three columns for price and market share in the US, EU, and UK. This is a critical error. In reality, geography is time. The sequence of regulatory approvals and reimbursement decisions across these markets directly dictates when cash flows arrive, materially altering an asset’s present value.

Across oncology programs approved from 2018 to 2022, for instance, FDA decisions consistently preceded those from the EMA, with a median lead time of approximately 300 days for new indications [29, 30]. This "US-first" phenomenon, driven by factors like earlier submissions and expedited review pathways, pulls the entire US revenue curve forward, compounding value year after year.

The dynamic in other key markets is distinct and must be modeled accordingly:

  • United Kingdom: While the MHRA's International Recognition Procedure can accelerate marketing authorization [31], the crucial metric for valuation is patient availability. This is dictated by HTA evaluation (NICE/SMC) and NHS commissioning, a process that historically takes a median of nearly 10 months from the authorization date [32]. This lag represents a significant delay in revenue generation.
  • European Union: Beyond regulatory review, national HTA and pricing negotiations introduce further delays, a pattern quantified by the EFPIA W.A.I.T. survey [34]. Processes like Germany’s AMNOG [3] and England’s Commercial Framework [4] create predictable calendars that push the start of meaningful revenue collection to the right.

The financial impact of these lags is not trivial. At a standard 10% discount rate, a 10-month delay in market access reduces the present value of that entire regional cash flow stream by over 7%.

Combining Price and Timing Disparities

When these temporal lags are combined with regional differences in net pricing, the impact on PV becomes even more pronounced. US net prices for pharmacy-benefit drugs typically reflect rebates of 35–55% [1], while physician-administered biologics sit closer to list [2]. In the EU and UK, confidential discounts and managed-entry agreements often result in net prices that are 20–40% or more below list [3, 4, 34].

This combined effect creates clear valuation patterns:

  • For specialty biologics, the combination of deeper EU/UK discounts and reimbursement lags means the present value per unit of volume can easily fall to half that of the US.
  • For cell and gene therapies, this disparity can be even wider. The high one-time payments are particularly sensitive to timing, and European managed access agreements often impose steep discounts to manage uncertainty.

A glass-box S0 replaces spreadsheet clones with a realistic, time-adjusted model. It understands that US approval is not the same as UK availability, and that an EU list price is not the same as a German net price. By embedding these legal and procedural calendars, you create a valuation that reflects how and when cash actually arrives.


4. Anchoring Valuation in Reality: Empirical Priors and Timelines

While S0 defines a program's potential commercial outcome, empirical priors provide the grammar of plausibility. They answer the fundamental questions that precede any financial projection: How likely is this program to succeed, and how long will it take? The most robust analyses from the past decade provide a clear and humbling baseline [20, 35, 36].

The Sobering Baseline for Clinical Success

A comprehensive analysis of clinical development from 2011–2020 established transition probabilities that should serve as the starting point for any evidence-based valuation [20]:

  • Phase I → Phase II: ~52%
  • Phase II → Phase III: ~28%
  • Phase III → Approval: ~56%
  • Overall Phase I → Approval: ~8%

These averages conceal important variations: oncology programs consistently underperform this baseline, while vaccines overperform. Biologics also generally demonstrate a higher overall likelihood of approval than small molecules. A pre-evidence valuation must be anchored to this landscape.

The Unyielding Development Calendar

Enthusiasm for "acceleration" often clashes with operational reality. Median clinical development durations have remained stubbornly long, reflecting the genuine friction of patient enrollment, data analysis, and regulatory interactions [21]:

  • Phase I: ~1.6 years
  • Phase II: ~2.9 years
  • Phase III: ~3.8 years

Valuations that compress these timelines without a concrete, detailed operational plan are merely substituting aspiration for analysis. The glass-box S0 built previously earns its credibility by aligning its commercial calendar with these empirical timelines.

Design, Not Optimism, Moves the Needle on Probability.

Priors are not destiny, but the basis from which improvement must be earned. The single most powerful lever for increasing the probability of success is the use of credible, prospectively defined patient selection biomarkers. In therapeutic areas like oncology, programs that rigorously enrich for responsive patients can roughly double their overall likelihood of approval, particularly at the crucial Phase II to Phase III transition [21]. This uplift is granted when the selection strategy is analytically validated and integral to the pivotal trial design.

A disciplined valuation process follows three practical steps. First, it explicitly writes these empirical priors and median timelines into its models. Second, it specifies which design features, such as a validated biomarker strategy, are permitted to move those priors to a higher posterior PoS. Third, it enforces governance that prevents S0 from being inflated until the evidence has crossed predefined thresholds.

Ultimately, S0 must remain tethered to these realities. Its value can only be justifiably "lifted" after the program’s posterior belief, its specific probability of success, has been improved through payer-credible evidence. Until then, S0's primary role is to act as a disciplined bound on enthusiasm.


5. Earning Credibility: The Role of Clinical Evidence Modeling and Bayesian Assurance

The simplest way to miscalculate an early-stage asset’s value is to confuse statistical power with the probability of success. Classical power answers a narrow, hypothetical question: "If the true effect is X, will this test detect it?" Governance requires a broader, more practical answer to the question: "Given our current beliefs and uncertainties about the effect, what is the actual chance this design will succeed?"

Bayesian Assurance is the method that provides this answer. It is the probability of success averaged over our current uncertainty, rather than being conditioned on a single, unknowable true effect. For this reason, Assurance is the appropriate metric for governing investment decisions at early gates.

Mapping Biology to Decision with Clinical Evidence Modeling (CEM)

Assurance is built upon a foundational layer: Clinical Evidence Modeling (CEM). CEM is a formalized process for mapping how early evidence components—such as target engagement, pharmacodynamic markers, or translational signals—predict the later endpoints that matter to regulators and payers [37, 38, 39]. This map quantifies the predictive strength of your early data, creating a transparent bridge from biology to a go/no-go decision.

When a therapeutic benefit is concentrated in a specific subpopulation, the design must reflect that. Predictive enrichment, prospectively defining and selecting the patients most likely to respond, can dramatically raise the probability of success and reduce required sample sizes. The high response rates seen with gefitinib in EGFR-mutant lung cancer and crizotinib in ALK-positive disease prove this principle: when mechanism and patient selection are aligned, design discipline delivers a powerful return on investment [40, 41].

Assurance as the Engine of Governance

In practice, an Assurance-based workflow brings auditability to the decision-making process. It involves:

  1. Clearly defining the trial's success criteria.
  2. Building a "prior" distribution that captures existing knowledge and uncertainty about the treatment effect. This can be derived from expert elicitation or historical data from similar programs [45, 51].
  3. Simulating the trial thousands of times under this uncertainty to calculate the prior predictive probability that the success rule will be met [42, 43].
  4. Stress-testing the result against key assumptions like sample size and enrichment strategy.

Assurance forces a shift from designing powerful trials based on optimistic assumptions to designing robust trials that have a high probability of success in the face of real-world uncertainty.

This methodology is particularly powerful where standard assumptions fail, such as in immuno-oncology trials with delayed effects [44]. More importantly, it aligns with how leadership teams think about risk. By setting clear Assurance targets for key gates (e.g., ≥40-60% for Proof-of-Concept, ≥60-70% for pivotal studies), organizations can create an objective, evidence-based system for advancing programs [46, 47].

Connecting Rigorous Design to Valuation

This brings the framework back to valuation. An early-stage asset's value is driven by its design-dependent Probability of Success, which Assurance provides. CEM and predictive enrichment offer the specific pathway to increase that probability through evidence. These probabilities then become direct inputs into dynamic valuation models like Real Options Analysis.

The governance output is powerful in its simplicity: invest when the Assurance for the next gate clears the organization’s threshold; wait or redesign when it does not; and divest when the posterior belief refuses to move with reasonable evidence. This discipline prevents enthusiasm from substituting for probability, keeping S0 in its proper role as a boundary condition while the real work of earning value through evidence unfolds.


6. Calibrating Risk by Modality: From Gene Therapy and Editing to ADCs

A program's valuation is quietly governed by its ability to navigate technical gates in manufacturing and comparability. The evidence frameworks discussed previously rely on priors and posterior probabilities of success (PoS), but these probabilities are not uniform across all therapeutic modalities. A rigorous model must adjust its PoS and timeline assumptions based on the specific risks inherent to each therapeutic class, explicitly referencing regulatory hurdles for manufacturing, comparability, and long-term safety [22, 23, 52].

When a program credibly passes these gates, a modest uplift in its posterior PoS is justified. When it cannot, discipline demands restraint.

Article content

Figure 4. Modality‑specific posterior probability of success (PoS) calibrations: from neutral priors to gate‑earned uplifts. Posterior PoS ranges across major therapeutic modalities—AAV in vivo, LNP‑mRNA, genome editing (in vivo), ex vivo HSC gene therapy, allogeneic cell therapy, small molecules, mAbs, and ADCs—conditioned on passage of CMC, comparability, and evidence gates. Each modality bar shows three bands: lower bound reflecting BIO 2011–2020 aggregate baselines; mid‑band achievable when design features (biomarker‑anchored enrichment, robust analytics, validated comparability) credibly reduce uncertainty; upper bound earned when programs demonstrate mature long‑term follow‑up data, tight σ control, and payer‑credible functional durability. AAV systemic: neutral priors sit below BIO baselines until genomic integrity, empty:full control, and complement immunosurveillance packages are complete; local AAV (retina, CNS) can access mid‑bands more readily. LNP‑mRNA: sits near BIO neutral when composition is locked and comparability is tight, reflecting mature manufacturing roadmaps and orthogonal potency validation. In vivo editing: starts conservatively below baselines, moves upward only after off‑target discovery, structural variant surveillance, and durable on‑target function are shown. Ex vivo HSC gene therapy: can exceed BIO baselines at Phase II→III transitions when multi‑year lineage‑specific pharmacodynamics are demonstrated, but σ and time multipliers remain elevated until potency comparability is established. Allogeneic cells: sit below autologous comparators until persistence or redose strategies are de‑risked. Small molecules follow BIO; mAbs trend slightly higher when PK/PD translation from non‑human primates is coherent; ADCs start below mAb priors until linker stability, multi‑analyte PK, and antigen biology are tightly matched. Ranges reflect regulatory expectations codified in FDA guidance on comparability, long‑term follow‑up, CAR‑T development, and ADC clinical pharmacology, overlaid on empirical transition probabilities from the decade‑level literature.

AAV Gene Therapy: The Challenge of Immunity and Manufacturing

Systemic AAV-delivered therapies face a repeating set of constraints that directly impact valuation. Pre-existing immunity limits the eligible patient pool, while immunogenic responses to the vector can trigger safety events. Furthermore, manufacturing platforms produce heterogeneous outputs, with variations in empty-to-full capsid ratios and impurity profiles creating uncertainty [55, 56, 57].

The valuation translation is straightforward: unless a program demonstrates control over these factors through robust analytics and a clear safety monitoring plan, it must start with a posterior PoS below industry baselines and assume wider schedule variance.

LNP-mRNA: The Power of Tractable Chemistry

In contrast, LNP-mRNA therapeutics offer a more predictable manufacturing and CMC profile, provided the core composition is fixed. The critical quality attributes—lipid chemistry, particle size, and mRNA modifications—are well-defined levers that control distribution, expression, and safety [53].

This tractability has a direct valuation counterpart. For LNP-mRNA programs with a locked composition and validated potency assays, PoS and timeline assumptions can reasonably align with standard industry benchmarks.

In Vivo Genome Editing: A Hybrid of High Reward and High-Risk Gates

In vivo editing programs combine the risks of their delivery vehicle (AAV or LNP) with the profound challenge of inducing a permanent genomic change. While the first proof-of-concept for LNP-CRISPR was a landmark achievement [54], it does not erase the high bar for evidence. Programs must demonstrate a lack of off-target effects, rule out major structural variants, and adhere to long-term patient follow-up requirements [23, 64].

The practical calibration is conservative by design. Initial PoS starts well below baselines, and timeline multipliers are increased to account for extensive genomic safety analysis. Posterior belief only moves toward mid-range bands after on-target editing, functional potency, and off-target safety are demonstrated with validated assays.

Ex Vivo and Allogeneic Cell Therapy: From Durability to Persistence

  • Ex vivo HSC Therapies: These programs pair the discipline of transplantation medicine with gene transfer analytics. Lentiviral vector-based therapies, for example, have demonstrated impressive durability, which can justify a higher PoS at the Phase II→III transition [58, 59]. However, this is balanced by significant operational complexities—patient conditioning, multi-week vein-to-vein times, and mandatory 15-year follow-up protocols—that keep timeline variance high [23, 60].
  • Allogeneic Therapies: "Off-the-shelf" cell products solve autologous manufacturing friction but introduce a new fundamental hurdle: host immune rejection and poor cell persistence [61, 62]. The evidentiary bar has shifted toward demonstrating durable benefit, which short-lived allogeneic platforms struggle to meet. Consequently, their PoS should be benchmarked below autologous comparators until persistence is credibly de-risked through engineering or redosing strategies.

ADCs and mAbs: Calibrating Risk in Established Modalities

Even classic modalities follow this architecture. Monoclonal antibodies (mAbs) often benefit from predictable pharmacokinetics, which can support tighter PoS estimates after early human data is available [66].

Antibody-drug conjugates (ADCs), however, add layers of complexity. Linker instability, drug-antibody ratio heterogeneity, and payload-related toxicities are common failure modes [65, 67, 68]. For valuation, this means an ADC program’s initial PoS should be set below that of a standard mAb. Uplift is earned only when these ADC-specific risks are addressed, antigen biology is well-understood, and conjugation quality is tightly controlled.

These modality-specific gates are not an alternative to the methods of CEM and Assurance; they are essential inputs. They define the realistic priors and variances within which those evidence-based frameworks can operate. A program's probability of success can only be as high as its modality-specific risks allow.


7. From Clinical Effect to Commercial Reality: Payer Thresholds and the Evidence Link Map

A therapy's commercial potential is ultimately defined by a "price-to-value corridor"—the space between what clinical science promises and what health systems can sustainably afford. A product does not earn routine use simply by demonstrating a clinical effect; it must prove that the magnitude and certainty of that effect justify a price that fits within constrained budgets and established care pathways.

The architecture of this corridor is not opaque. It is codified in the public methodologies of HTA bodies like NICE in England [11], IQWiG in Germany [14], and reflected in the value frameworks of organizations like ICER in the U.S. [23]. These frameworks reveal that a therapy’s price and adoption speed are governed by three primary forces.

The Three Forces Shaping Price and Access

  1. Severity: Health systems explicitly value treatments for severe conditions with high unmet need. Frameworks like NICE’s Highly Specialized Technologies (HST) program apply weighting that allows for higher cost-effectiveness ranges, effectively lifting the price corridor for therapies targeting these diseases [11]. The greater and more certain the benefit over the standard of care, the stronger the negotiating position.
  2. Uncertainty: Payers discount uncertainty. A premium is paid for mature, pivotal evidence demonstrating a clear benefit on endpoints that matter, like overall survival. When evidence is less mature (e.g., based on surrogate endpoints), payers respond with managed access schemes, coverage with evidence development, or confidential discounts to mitigate risk [4, 11]. The price corridor tightens when uncertainty is high.
  3. Affordability: Even when a therapy is deemed cost-effective, its adoption rate is constrained by budget impact and operational realities. NICE’s resource-impact templates, which project gradual uptake for chronic therapies over several years (e.g., 20% to 50% from Y1-Y3), are not arbitrary guesses but reflections of clinic capacity and financial envelopes [14]. In the US, utilization management tools like prior authorization serve the same function, slowing penetration for drugs priced outside accepted value benchmarks.

The Evidence Link Map: A Tool for Strategic Development

The Evidence Link Map is a structured translation layer that connects a developer’s strategic choices to the outcomes that payers recognize as value. It makes explicit how specific development activities can influence price and access.

Key Levers to Influence Payer Value:

  • Biomarker Selection: Concentrating a therapeutic benefit in a defined population can justify a higher price and faster penetration—but only if the selection rule is prospectively validated and testing is operationally feasible.
  • Endpoint Maturation: Confirming an early surrogate signal with mature overall survival data reduces uncertainty and can unlock higher price bands, particularly in high-severity contexts [11].
  • Evidence Generation: Using Real-World Evidence (RWE) or outcomes-based agreements to address residual uncertainty (e.g., long-term durability) can enable a transition from restricted, managed access to routine commissioning [4, 11, 23].

These principles create clear "if-then" patterns. If overall survival data is immature, expect managed access with an evidence-generation commitment. If a predictive biomarker is required, adoption becomes a function of testing penetration. This framework stops the pretense that a clinical effect automatically translates to market adoption.

Article content

Figure 5. The Evidence Link Map: translating clinical effect into payer‑credible price corridors and adoption trajectories. Structured translation layer connecting development levers (endpoint choice, effect size, evidence maturity, biomarker selection, RWE augmentation, outcomes‑based arrangements) to HTA and payer parameters for price corridors and access pathways. Three primary modifiers shape corridors: Severity lifts corridors when life expectancy losses are large and unmet need is clear (NICE HST QALY weighting, IQWiG added‑benefit categories). Uncertainty tightens corridors when evidence is immature or surrogate; widens when OS matures, comparative effectiveness is demonstrated, and durability is shown. Affordability gates adoption speed via budget constraints, commissioning capacity, and operational readiness (NICE resource‑impact profiles: 20%, 35%, 50% over Years 1–3 for chronic therapies). Development levers: Biomarker‑anchored selection moves effect size and penetration (contingent on testing infrastructure). OS maturation reduces uncertainty, unlocks movement within HST or ICER benchmark corridors. RWE commitments answering residual questions (durability, safety in excluded populations, comparative effectiveness) relax managed access gating. Outcomes‑based arrangements enable earlier access at higher net prices by insuring payers against overpayment. If‑then patterns: immature OS → managed access + registry commitment; predictive biomarker required → adoption = testing penetration × adherence; specialized delivery sites → capacity caps until activation matures. Anchored to HTA frameworks: NICE methods (severity modifiers, cost‑effectiveness ranges), ICER ($100k–$150k/QALY benchmarks), IQWiG (added‑benefit tied to evidence strength), HAS (economic evaluation), England Commercial Framework (managed access, confidential discounts).

From a governance perspective, this ties back to the central thesis: S0 is only permitted to move when the evidence becomes payer-credible. The price corridors and adoption curves in our glass-box model must reflect these HTA-driven realities. Only when a program generates evidence that reduces uncertainty and aligns with what payers value—using levers from the Evidence Link Map—does the model have permission to lift S0 toward more optimistic scenarios.


8. Valuing the Deal: Layering Platform Options onto a Sober S0

A strategic partnership is an option tree, where each branch represents an opportunity for a partner to invest only after uncertainty has been reduced. For a company with a true asset-generation engine, its valuation extends beyond the base value of its named assets (Layer A). Layer B captures the economics of the platform itself: the option fees, milestone payments, and royalty streams that partners pay for access to this engine.

Misinterpreting these deal structures leads to inflated valuations built on headline figures. A disciplined approach, however, models them as a series of timed, probability-weighted cash flows that are layered onto the foundational S0.

The Anatomy of a Platform Deal

Publicly announced collaborations provide a clear blueprint for how these option-like terms are structured:

  • Baskets and Expansion Rights: A multi-program deal signals engine throughput. Agreements like Recursion’s with Roche/Genentech (up to 40 programs) or Generate Biomedicines' with Amgen (5 initial targets with an option for 5 more) anchor per-program economics while creating future monetization events for the platform [69, 76].
  • Gates and Opt-in Fees: A partner’s decision to exercise an option is a powerful valuation signal. These events provide real-world data on what a specific piece of evidence is worth. Gilead’s $20 million option exercise for a Nurix degrader [72, 73] and Novartis’s $15 million exercise for a Voyager AAV capsid [74, 75] serve as crucial calibration points for fee size and timing at key development gates.
  • Milestone Ladders and Royalty Tiers: Total deal values are misleading. The value lies in the per-program structure. AI discovery collaborations often feature potential milestones of $300–$400 million per program with royalties in the high-single to mid-teen percentages [69, 70]. In contrast, enabling technology licenses, like for AAV capsids, command lower economics, with smaller milestones and mid- to high-single-digit royalties [74, 75].
  • Profit-Share Toggles: The right to co-invest and share profits (e.g., a 50/50 US split) represents a high-conviction option. It increases the potential return but also the financial risk at a critical moment in development, a structure seen in deals from Kymera [71] and Life Edit [78].

The Discipline for Modeling Platform Value

A credible platform valuation avoids double-counting by adhering to a strict process. Never simply aggregate the "up to $X billion" headline values. Instead, you must:

  1. Parameterize the milestone ladder for each program into research, regulatory, and commercial payments.
  2. Probability-weight each gate based on empirical success rates and data from public option exercises.
  3. Layer these timed cash flows (inflows like fees and milestones; outflows like royalties paid on a licensed component) on top of the base S0 (Layer A) for each asset.

This layered approach recognizes that you do not own the headline value. You own the ability to generate evidence that persuades a partner to exercise the next option. That persuasion is built from the same machinery assembled in previous sections: Clinical Evidence Modeling, Bayesian Assurance, modality-specific risk mitigation, and a payer-focused Evidence Link Map.

The options in Layer B are only exercised when the science in Layer A is sound.


9. The Framework in Action: Micro-Examples and Parameter Packs

These brief examples are designed to make the valuation machinery tangible. Each scenario uses a conservative parameter pack, grounded in empirical data, to demonstrate how the core principles—a transparent S0, design-dependent probabilities, discontinuous risk, the finite horizon, and platform overlays—function in practice.

A) The Credible S0: A Realistic Base Case

A defensible S0 is an assembly of traceable assumptions. For a chronic, pharmacy-benefit therapy, this includes:

  • Price Corridors: US net prices reflect average rebates of 35-55% off list price [1], while EU/UK net prices are typically 20-40% below list due to managed access agreements [3, 4].
  • Adoption Archetypes: Uptake is not a generic curve. It follows patterns dictated by budget and clinical capacity, such as the 20-50% three-year ramp seen in some NICE resource templates [14].
  • Time-Adjusted Geographies: Launch sequences reflect reality, with US approvals often preceding the EU by months and UK availability lagging marketing authorisation significantly [29, 31, 32, 34].
  • LOE Dynamics: Market erosion is modeled based on modality, from the rapid price collapse of small molecules following generic entry [11] to the more gradual, but still steep, decline of biologics.

An S0 built this way provides a set of justifiable dials, not unsubstantiated promises.

B) Posterior PoS: The True Fulcrum of Early-Stage Value

An asset’s early value is driven by its probability of success (PoS), not S0. The starting point is sober priors: the decade-long average for a drug moving from Phase I to approval is only ~8% [20].

Uplift from this baseline is not achieved by assertion, but is earned through superior design and governance. This is true from the very beginning. At the preclinical stage, value is created not by claims of a "best-in-class target" but by instituting disciplined go/no-go decision gates and robust project management to navigate the high rate of attrition.

In the clinic, the most powerful lever is a prospectively validated biomarker, which can nearly double the overall likelihood of approval [21]. Methods like Clinical Evidence Modeling and Bayesian Assurance provide the formal machinery to translate these design choices into a credible posterior PoS. This updated probability, not the S0, is what truly moves the needle on early-stage valuation.

C) Discontinuous Risk: Modeling Cliffs, Not Fog

Uncertainty in biotech is not a smooth drift; it is a series of high-impact, discontinuous "jumps" at key gates. These include IND-enabling toxicology results, Phase II data readouts, pivotal trial outcomes, and major reimbursement decisions. An honest risk model places these jump nodes at the discrete moments where a program’s fate is actually decided, rather than pretending risk is evenly distributed over time. This brings discipline by focusing attention and capital on navigating these make-or-break events.

D) q: The Quantifiable Cost of Delay

The cost of waiting is a calculable "convenience yield" (q) that erodes value over time. It is a composite of three forces:

  1. q_patent: The value lost to a shrinking patent runway [24, 25].
  2. q_comp: The penalty for arriving late in a competitive therapeutic area.
  3. q_payer: The risk of facing stricter evidence requirements or price negotiation regimes over time [26, 27, 28].

By quantifying q, the cost of every operational delay becomes explicit, forcing a disciplined approach to managing development timelines.

E) Geography as Present Value: Calendars Are Cash

As established, geography is time. The calendar of regional approvals and reimbursement decisions directly impacts present value. A model that fails to build in the typical months-long lags between FDA approval [29] and funded availability in markets like the UK [32] will materially overstate an asset’s worth. A rigorous valuation codifies these offsets as default settings.

F) Platform Overlays: Cashing in on Information

Layer B platform value is found in the public record of deal-making. Option exercise fees ($15-20M at candidate selection [72, 73, 74, 75]), per-program milestone ladders ($300-400M in AI discovery deals [69, 70, 76]), and profit-share toggles [70, 71, 78] are anchors for what a partner will pay when evidence reduces uncertainty. A disciplined model layers these probability-weighted cash flows on top of Layer A, using disclosed deal terms as the basis for valuation.

Putting It All Together: The relative importance of these drivers changes by asset type. An early-stage oncology small molecule is dominated by its posterior PoS and the high cost of delay (q). A rare disease gene therapy is constrained by modality-specific gates and wider payer corridors. A late-stage immunology mAb is driven heavily by the geographic and commercial texture of its S0. True platform value (Layer B) is then layered on top of any of these scenarios, reflecting timed, risk-adjusted cash flows from strategic partnerships.


10. Synthesis and Conclusions

A single valuation number is a convenient fiction. It obscures the levers that ultimately determine a program's fate. The architecture I have detailed replaces this simplicity with a working system for making auditable decisions. It comprises a transparent "glass-box" S0, empirical probabilities of success, a model of discontinuous risk, and a clear accounting for the costs of time, geography, and payer demands.

What you now possess is a framework to answer not just "what is it worth?" but "why is it worth that?"

Key Pillars of a Disciplined Valuation Architecture

  • The Glass-Box S0: This is the foundation. By deconstructing the commercial forecast into its component parts (auditable price corridors, adoption archetypes, and LOE schedules) we anchor the model in sourced evidence. Disagreements become tractable debates about parameters, not positions. This S0 sets a disciplined bound on optimism.
  • Belief Earned Through Evidence: Early-stage value is not driven by S0. It is driven by the posterior probability of success (PoS). This starts with sober industry priors (~8% from Phase I to approval) and is improved through superior program design. At the preclinical stage, uplift is earned via operational rigor and disciplined go/no-go governance. In the clinic, methods like Clinical Evidence Modeling and Bayesian Assurance provide the formal mathematics to quantify how design choices, such as using a validated biomarker, translate into a higher posterior PoS.
  • Risk That Arrives in Jumps: Uncertainty is not evenly distributed. It concentrates at discrete, high-impact gates: IND-enabling tox results, Phase II readouts, pivotal outcomes, and HTA decisions. Modeling this "jump risk" forces an honest confrontation with where capital is truly at risk and where decisions actually hinge.
  • Time as a Tangible Cost: Delay is not a soft cost; it is a calculable loss of value. By quantifying the convenience yield (q), comprising patent erosion, competitive pressure, and payer tightening, we make the price of every timeline slip explicit.
  • The Commercial Realities of Geography and Payers: Present value is shaped by when and where cash arrives. Modeling the real-world lags between regulatory approval and funded access across the US, EU, and UK is non-negotiable. Furthermore, the Evidence Link Map connects clinical effect sizes to actual price and penetration by acknowledging that payers reward certainty and demonstrated value on endpoints that matter.
  • Platform Overlays as Timed, Risk-Adjusted Cash: True platform value (Layer B) is not an abstract premium. It is a series of option-like cash flows from partnerships, which are exercised only when the underlying science (Layer A) is de-risked. A disciplined model layers these probability-weighted fees, milestones, and royalties on top of the asset-level valuation.

Three Core Governance Outputs

This architecture produces three tangible outputs for decision-making rooms:

  1. Invest–Wait–Kill Thresholds: By setting and enforcing objective Assurance targets for each development gate (e.g., 40-60% for PoC; 60-70% for pivotal studies), organizations can stop funding false positives based on enthusiasm.
  2. The Option Discipline for Time: By quantifying the cost of delay (q), the value of operational excellence becomes visible, justifying investments that protect or accelerate the development clock.
  3. An Auditable Artifact Bundle: The glass-box S0, Evidence Link Map, and parameter pack transform valuation from a subjective narrative into a durable, transferable system, ensuring decisions are grounded in a consistent, evidence-based framework.

S0 can serve as a savior or a boundary. The discipline outlined here ensures it functions as the latter, keeping ambition tethered to evidence until the data has truly earned the right to lift value.


This is a summarized version of an in-depth article published on our website. For a more comprehensive exploration with expanded sections and case studies visit "Building Biotech Valuations That Survive Reality: Priors, Evidence, and Finite Horizons". Special thanks to Anabel Perez-Gomez for critical reading of the manuscript and valuable insights.

Building glass-box S0 models, computing design-dependent Assurance, and structuring platform valuations requires a rigorous framework that connects strategy and evidence to payer realities and capital decisions. At INBISTRA, we help biotech companies and investors implement these methods to build auditable valuation architectures and strengthen investment governance.


References

[1] Congressional Budget Office (CBO). Prescription Drugs: Spending, Use, and Prices. January 2022. https://www.cbo.gov/system/files/2022-01/57050-Rx-Spending.pdf

[2] Medicare Payment Advisory Commission (MedPAC). Addressing the high prices of drugs covered under Medicare Part B. March 2, 2023. https://www.medpac.gov/wp-content/uploads/2022/07/Part-B-drugs-March-2023-SEC.pdf

[3] Gemeinsamer Bundesausschuss (G‑BA). Benefit Assessment of Medicinal Products (AMNOG) — responsibilities and pricing procedure. Accessed Oct 2025. https://www.g-ba.de/english/benefitassessment/

[4] NHS England. NHS Commercial Framework for New Medicines (updated Jan 2025). https://www.england.nhs.uk/medicines-2/medicines-value-and-access/nhs-commercial-framework-for-new-medicines/

[5] Vogler S, Schneider P, Zimmermann N, Habl C, Piessnegger J. Medicine price transparency and confidential managed-entry agreements in Europe: findings from the EURIPID survey. Health Policy. 2021. https://pubmed.ncbi.nlm.nih.gov/34253396/

[6] Institute for Clinical and Economic Review (ICER). Obesity Management (Semaglutide 2.4 mg) — Report-at-a-Glance and Final Evidence Report. 2022. https://icer.org/assessment/obesity-management-2022/

[7] National Institute for Health and Care Excellence (NICE). TA733: Inclisiran for treating primary hypercholesterolaemia or mixed dyslipidaemia. 6 Oct 2021. https://www.nice.org.uk/guidance/ta733

[8] NICE. HST15: Onasemnogene abeparvovec (Zolgensma) for treating spinal muscular atrophy. 2021 (updated 2023). https://www.nice.org.uk/guidance/hst15

[9] NICE. TA872: Axicabtagene ciloleucel for DLBCL/PMBCL after ≥2 lines — 28 Feb 2023. https://www.nice.org.uk/guidance/ta872

[10] Amgen. Amgen Makes Repatha Available in the US at a 60 Percent Reduced List Price. Press release, 24 Oct 2018. https://www.amgen.com/newsroom/press-releases/2018/10/amgen-makes-repatha-evolocumab-available-in-the-us-at-a-60-percent-reduced-list-price

[11] U.S. Food and Drug Administration (FDA). Generic Competition and Drug Prices: New Evidence Linking Greater Generic Competition and Lower Generic Drug Prices. December 2019. https://www.fda.gov/media/133509/download

[12] Office of the Assistant Secretary for Planning and Evaluation (ASPE), HHS. Medicare Part B Enrollee Use and Spending on Biosimilars, 2018–2023. January 2025. https://aspe.hhs.gov/sites/default/files/documents/be065dbbd1f866c65cf627995bd2ea56/biosimilars-medicare-part-b.pdf

[13] AbbVie. AbbVie Reports Full‑Year and Fourth‑Quarter 2023 Financial Results. 2 Feb 2024. https://news.abbvie.com/2024-02-02-AbbVie-Reports-Full-Year-and-Fourth-Quarter-2023-Financial-Results

[14] NICE. TA679 Resource Impact Report: Dapagliflozin for treating chronic heart failure with reduced ejection fraction. Mar 2021; updated Mar 2022. https://www.nice.org.uk/guidance/ta679/resources/resource-impact-report-pdf-9021358765

[15] FDA. FDA approves osimertinib for first‑line treatment of metastatic NSCLC with common EGFR mutations. 18 Apr 2018. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-approves-osimertinib-first-line-treatment-metastatic-nsclc-most-common-egfr-mutations

[16] FDA. FDA grants accelerated approval to sotorasib for KRAS G12C‑mutated NSCLC. 28 May 2021. https://www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-sotorasib-kras-g12c-mutated-nsclc

[17] NICE. TA781: Sotorasib for previously treated KRAS G12C mutation‑positive advanced NSCLC — Resource impact statement. 30 Mar 2022. https://www.nice.org.uk/guidance/ta781/resources/resource-impact-statement-11015109613

[18] AstraZeneca. Annual Report 2019 — Oncology highlights (Tagrisso). 2019. https://www.astrazeneca.com/investor-relations/annual-reports/annual-report-2019.html

[19] FDA. Approved Drug Products (Orange Book) — patents/exclusivity FAQs. Accessed Oct 2025. https://www.fda.gov/drugs/drug-approvals-and-databases/approved-drug-products-therapeutic-equivalence-evaluations-orange-book

[20] Biotechnology Innovation Organization (BIO); QLS Advisors; Informa Pharma Intelligence. Clinical Development Success Rates 2011–2020. 2021. PDF

[21] Wong C‑H, Siah KW, Lo AW. Estimation of clinical trial success rates and related parameters. Biostatistics. 2019;20(2):273–286. PMID: 29394327. https://pubmed.ncbi.nlm.nih.gov/29394327/

[22] U.S. FDA, CBER. Manufacturing Changes and Comparability for Human Cellular and Gene Therapy Products (Draft Guidance). July 2023. https://www.fda.gov/media/170198/download

[23] U.S. FDA, CBER. Long Term Follow‑Up After Administration of Human Gene Therapy Products (Final Guidance). January 2020. https://www.fda.gov/files/vaccines%2C%20blood%20%26%20biologics/published/Long-Term-Follow-Up-After-Admin-Human-GT-Products_Jan_2020.pdf

[24] U.S. Patent and Trademark Office (USPTO). MPEP §2758 — Calculation of Patent Term Extension (35 U.S.C. §156). Accessed Oct 2025. https://www.uspto.gov/web/offices/pac/mpep/s2758.html

[25] European Commission. Supplementary protection certificates for pharmaceutical products (SPCs). Accessed Oct 2025. Link

[26] KFF (Kaiser Family Foundation). FAQs about the Inflation Reduction Act’s Medicare Drug Price Negotiation Program. 2025. https://www.kff.org/medicare/faqs-about-the-inflation-reduction-acts-medicare-drug-price-negotiation-program/

[27] Centers for Medicare & Medicaid Services (CMS). Medicare Drug Price Negotiation Program — 2027 Cycle: Final Guidance and MFP Effectuation (Fact Sheet). 2025. Link

[28] European Commission. Joint Clinical Assessments under the EU HTA Regulation (EU) 2021/2282 — Implementation Page. Accessed Oct 2025. Link

[29] Cramer A, Sørup FKH, Christensen HR, Petersen TS, Karstoft K. Cancer drug applications to the EMA and the FDA: A comparison of new drugs and extension of indication in terms of approval decisions and time in review. Br J Clin Pharmacol. 2025;91(5):1431–1438. https://pmc.ncbi.nlm.nih.gov/articles/PMC12035586/

[30] U.S. Food and Drug Administration (FDA). Priority Review. Accessed 2025. https://www.fda.gov/patients/fast-track-breakthrough-therapy-accelerated-approval-priority-review/priority-review

[31] Medicines and Healthcare products Regulatory Agency (MHRA). International Recognition Procedure (IRP). GOV.UK. 2024. https://www.gov.uk/government/publications/international-recognition-procedure/international-recognition-procedure

[32] Office for Life Sciences (UK). Life sciences competitiveness indicators 2024 — Summary. GOV.UK. 2024. https://www.gov.uk/government/publications/life-sciences-sector-data-2024/life-sciences-competitiveness-indicators-2024-summary

[33] Scottish Medicines Consortium. Timelines & Submission Scheduling. Accessed 2025. https://scottishmedicines.org.uk/making-a-submission/timelines-submission-scheduling/

[34] EFPIA. Patients W.A.I.T. Indicator 2024 Survey (Final Report, Apr 2025). https://efpia.eu/media/oeganukm/efpia-patients-wait-indicator-2024-final-110425.pdf

[35] Hay M, Thomas DW, Craighead JL, Economides C, Rosenthal J. Clinical development success rates for investigational drugs. Nature Biotechnology. 2014;32(1):40–51. https://doi.org/10.1038/nbt.2786

[36] Smietana K, Siatkowski M, Møller M. Trends in clinical success rates. Nature Reviews Drug Discovery. 2016;15(6):379–380. https://doi.org/10.1038/nrd.2016.85

[37] U.S. Food and Drug Administration (FDA). Enrichment Strategies for Clinical Trials to Support Determination of Effectiveness of Human Drugs and Biological Products (Guidance for Industry). March 2019. https://www.fda.gov/media/121320/download

[38] Mandrekar SJ, Sargent DJ. Clinical trial designs for predictive biomarker validation: enrichment, all-comers, and adaptive strategies. Journal of Clinical Oncology. 2013. PMID: 23665336. https://pubmed.ncbi.nlm.nih.gov/23665336/

[39] Evrard YA, et al. Incorporating molecular diagnostics into early clinical trials and the ET-CTN framework. 2015. PMID: 26423340. https://pubmed.ncbi.nlm.nih.gov/26423340/

[40] Crizotinib in ALK-positive NSCLC: Phase I/II program summary. Drugs. 2011. PMID: 22191798. https://pubmed.ncbi.nlm.nih.gov/22191798/

[41] Gefitinib for chemotherapy‑naïve, EGFR‑mutated advanced NSCLC: Phase II study. 2006. PMID: 16785471. https://pubmed.ncbi.nlm.nih.gov/16785471/

[42] Assurance for survival outcomes; prior elicitation and software. 2013. PMID: 23861270. https://pubmed.ncbi.nlm.nih.gov/23861270/

[43] Eliciting a distribution for a variance parameter and incorporating it in Assurance; two‑stage development. 2020. PMID: 32537910. https://pubmed.ncbi.nlm.nih.gov/32537910/

[44] Assurance for trials with delayed treatment effects in immuno‑oncology; elicitation and weighted testing. 2024. PMID: 38881219. https://pubmed.ncbi.nlm.nih.gov/38881219/

[45] O’Hagan A, et al. Multivariate expert elicitation within SHELF; program‑level PoS. 2022. PMID: 35373454. https://pubmed.ncbi.nlm.nih.gov/35373454/

[46] Bayesian development simulation in rheumatoid arthritis; program‑level Assurance. 2009. PMID: 19340851. https://pubmed.ncbi.nlm.nih.gov/19340851/

[47] Assurance adoption and case studies in industry; decomposition into conditional PoS. 2018. PMID: 29635777. https://pubmed.ncbi.nlm.nih.gov/29635777/

[48] Vaccine development: using immunological endpoints to form priors for Phase 3 efficacy Assurance. 2021. PMID: 34254347. https://pubmed.ncbi.nlm.nih.gov/34254347/

[49] Probability of success post‑interim (PoS|Continue) in group sequential trials. 2024. PMID: 39648488. https://pubmed.ncbi.nlm.nih.gov/39648488/

[50] Selecting the design effect by equating frequentist power to conditional expected power. 2015. PMID: 25583273. https://pubmed.ncbi.nlm.nih.gov/25583273/

[51] Schmidli H, Gsteiger S, Roychoudhury S, O’Hagan A, Spiegelhalter D, Neuenschwander B. Robust meta-analytic-predictive priors in clinical trials with historical control information. Biometrics. 2014;70(4):1023–1032. https://pubmed.ncbi.nlm.nih.gov/25355546/

[52] International Council for Harmonisation (ICH). Q5E: Comparability of Biotechnological/Biological Products Subject to Changes in Their Manufacturing Process. 2004. https://www.ich.org/page/quality-guidelines

[53] Hou X, Zaks T, Langer R, Dong Y. Lipid nanoparticles for mRNA delivery. Nature Reviews Materials. 2021;6:1078–1094. PMID: 34394960. https://pubmed.ncbi.nlm.nih.gov/34394960/

[54] Gillmore JD, Gane E, Taubel J, et al. CRISPR–Cas9 In Vivo Gene Editing for Transthyretin Amyloidosis. New England Journal of Medicine. 2021;385:493–502. PMID: 34215024. https://pubmed.ncbi.nlm.nih.gov/34215024/

[55] Shen W, Liu S, Ou L. rAAV immunogenicity, toxicity, and durability in 255 clinical trials: A meta-analysis. Frontiers in Immunology. 2022;13:1001263. https://pubmed.ncbi.nlm.nih.gov/36389770/

[56] Monahan PE, Négrier C, Tarantino M, Valentino LA, Mingozzi F. Emerging immunogenicity and genotoxicity considerations of adeno-associated virus vector gene therapy for hemophilia. Journal of Clinical Medicine. 2021;10(11):2471. https://pubmed.ncbi.nlm.nih.gov/34199563/

[57] Brimble MA, Winston SM, Davidoff AM. Stowaways in the cargo: Contaminating nucleic acids in rAAV preparations for gene therapy. Molecular Therapy. 2023;31(10):2826–2838. https://pubmed.ncbi.nlm.nih.gov/37533254/

[58] De Ravin SS, Malech HL, et al. Hematopoietic gene therapy: progress toward safe and effective treatments. Molecular Therapy. 2019;27(5):895–906. https://pubmed.ncbi.nlm.nih.gov/31231794/

[59] Kohn DB, Candotti F, Thrasher AJ, et al. Gene therapy for primary immune deficiencies: state of the art. Current Opinion in Allergy and Clinical Immunology. 2021;21(6):545–556. https://pubmed.ncbi.nlm.nih.gov/33717203/

[60] Park SH, et al. Hematopoietic stem cell gene therapy/editing for sickle cell disease: strategies and status. International Journal of Molecular Sciences. 2022;23(11):6038. https://pubmed.ncbi.nlm.nih.gov/35681538/

[61] Larson RC, Maus MV. Recent advances and discoveries in the mechanisms and functions of CAR T cells. Nature Reviews Cancer. 2021;21(3):145–161. https://pubmed.ncbi.nlm.nih.gov/33483715/

[62] Khawar MB, Sun H. CAR-NK Cells: From Natural Basis to Design for Kill. Frontiers in Immunology. 2021;12:707542. https://pubmed.ncbi.nlm.nih.gov/34970253/

[63] Lee DW, Santomasso BD, Locke FL, et al. ASTCT Consensus Grading for Cytokine Release Syndrome and Neurologic Toxicity Associated with Immune Effector Cells. Biology of Blood and Marrow Transplantation. 2019;25(4):625–638. https://pubmed.ncbi.nlm.nih.gov/30592986/

[64] U.S. FDA, CBER. Human Gene Therapy Products Incorporating Human Genome Editing (Guidance for Industry). 2024. https://www.fda.gov/media/156894/download

[65] U.S. FDA, CDER. Clinical Pharmacology Considerations for Antibody‑Drug Conjugates. March 2024. https://www.fda.gov/media/177250/download

[66] Liu L. Pharmacokinetics of monoclonal antibodies and Fc-fusion proteins. Protein & Cell. 2018;9(1):15–32. https://pubmed.ncbi.nlm.nih.gov/28421387/

[67] Kraynov E, Kamath AV, Walles M, et al. Current approaches for absorption, distribution, metabolism, and excretion characterization of antibody-drug conjugates: An industry white paper. Drug Metabolism and Disposition. 2016;44(5):617–623. https://pubmed.ncbi.nlm.nih.gov/26669328/

[68] Singh AP, Shin YG, Shah DK. Application of pharmacokinetic-pharmacodynamic modeling and simulation for antibody-drug conjugate development. Pharmaceutical Research. 2015;32(11):3508–3525. https://pubmed.ncbi.nlm.nih.gov/25666843/

[69] Recursion Pharmaceuticals. Recursion Announces Transformational Collaboration with Roche and Genentech. Press release. 7 Dec 2021. https://ir.recursion.com/news-releases/news-release-details/recursion-announces-transformational-collaboration-roche-and

[70] Sanofi. Exscientia and Sanofi establish strategic research collaboration to develop AI‑driven pipeline. Press release. 7 Jan 2022. https://www.sanofi.com/en/media-room/press-releases/2022/2022-01-07-06-00-00-2362917

[71] Kymera Therapeutics. Kymera Therapeutics and Sanofi Enter into Strategic Partnership to Advance Novel Protein Degrader Therapies. Press release. 9 Jul 2020. https://investors.kymeratx.com/news-releases/news-release-details/kymera-therapeutics-and-sanofi-enter-strategic-partnership

[72] Gilead Sciences. Gilead and Nurix Establish Strategic Collaboration to Develop Novel Therapies. Press release. 19 Jun 2019. https://www.gilead.com/news/news-details/2019/gilead-and-nurix-establish-strategic-collaboration-to-develop-novel-therapies-for-cancer-and-other-diseases

[73] Nurix Therapeutics. Gilead Exercises Option to License Nurix’s IRAK4 Targeted Protein Degrader NX‑0479 (GS‑6791). Press release. 20 Mar 2023. https://ir.nurixtx.com/news-releases/news-release-details/gilead-exercises-option-license-nurixs-irak4-targeted-protein

[74] Novartis. Novartis, Voyager Therapeutics reach license option agreement for next‑generation gene therapy vectors for neurological diseases. Press release. 8 Mar 2022. https://www.novartis.com/news/media-releases/novartis-voyager-therapeutics-reach-license-option-agreement-next-generation-gene-therapy-vectors-neurological-diseases

[75] Voyager Therapeutics. Voyager Enters into License for Next‑Generation Capsid (Novartis). Press release. 5 Sept 2024. https://ir.voyagertherapeutics.com/news-releases/news-release-details/voyager-enters-license-next-generation-capsid-bringing-partnered

[76] Amgen; Generate Biomedicines. Amgen and Generate Biomedicines Announce Multi‑Target, Multi‑Modality Research Collaboration Agreement. Press release (PR Newswire). 6 Jan 2022. https://www.prnewswire.com/news-releases/amgen-and-generate-biomedicines-announce-multi-target-multi-modality-research-collaboration-agreement-301455157.html

[77] Scribe Therapeutics. Scribe Therapeutics Announces Research Collaboration with Sanofi to Accelerate CRISPR‑based Cell Therapies for Cancer. Business Wire press release. 27 Sept 2022. https://www.businesswire.com/news/home/20220927005568/en/Scribe-Therapeutics-Announces-Research-Collaboration-with-Sanofi-to-Accelerate-Breakthrough-CRISPR-based-Cell-Therapies-for-Cancer

[78] ElevateBio / Life Edit Therapeutics. Novo Nordisk and Life Edit Therapeutics Establish Multi‑Target Collaboration to Discover and Develop Gene Editing Therapies for Rare and Cardiometabolic Diseases. Company press release. 24 May 2023. https://elevate.bio/press-releases/novo-nordisk-and-life-edit-therapeutics-establish-multi-target-collaboration-to-discover-and-develop-gene-editing-therapies-for-rare-and-cardiometabolic-diseases/


To view or add a comment, sign in

More articles by Ignacio Sancho-Martinez, PhD

Explore content categories