Language models can write brilliant analytical reports, but produce different answers every time. We fixed that with math borrowed from Maxwell’s equations.
Listen to the article on Apple Podcasts
Listen to the article on Soundcloud
Related Article: Flat Facts, Curved Beliefs: A Geometric Hypothesis for Transformer Cognition
Related Article: We Found Something Strange When We Connected Two AI Minds
Abstract
Large language models demonstrate remarkable capability in technical writing but exhibit fundamental non-determinism that renders them incompatible with regulatory requirements in pharmaceutical manufacturing. When generating Good Manufacturing Practice (GMP) analytical reports, state-of-the-art models produce inconsistent outputs across identical inputs — a critical failure mode in environments governed by FDA data integrity standards (21 CFR Part 11, ALCOA+ principles).
We introduce cognitive anchoring, a gauge-theoretic framework that synchronizes multi-headed attention in transformer architectures through the use of structured invariants. Applied to field equation discovery tasks, cognitive anchoring achieves 38% improvement in symbolic consistency (embedding similarity: 0.94 vs. 0.68) and 31% reduction in minimum description length. We demonstrate how these principles extend to pharmaceutical analytical testing, where deterministic reasoning enables the adoption of AI in regulated environments for the first time. This work establishes a foundation for trustworthy AI in industries where reproducibility is not optional but mandatory.
Introduction
The pharmaceutical industry faces a paradox. Artificial intelligence promises to transform analytical testing workflows — accelerating report generation, reducing transcription errors, and freeing PhD-level scientists from hours of manual documentation. Yet regulatory frameworks designed to ensure drug safety demand absolute reproducibility in documentation practices. The FDA’s data integrity guidance requires that analytical reports demonstrate attributability, legibility, contemporaneousness, originality, and accuracy (ALCOA+). European Medicines Agency (EMA) standards mandate complete audit trails and version control. These requirements assume deterministic processes: identical inputs must yield similar outputs.
Current large language models violate this fundamental assumption. When GPT-4 is asked five times to generate a sterility testing report from identical instrument data, it produces five structurally different documents — varying in section order, terminology choices, statistical interpretations, and regulatory references. This non-determinism is not an implementation bug but an architectural feature: transformer models employ stochastic sampling during inference, and parallel attention heads pursue independent optimization objectives without explicit coordination.
The consequences extend beyond technical inconvenience. Non-reproducible AI systems cannot be validated under Good Manufacturing Practice standards. They introduce audit risk, as regulators flag documentation inconsistency as evidence of inadequate process control. They undermine the value proposition of automation — if human QA scientists must extensively revise each AI-generated report to ensure consistency, efficiency gains disappear.
This paper addresses a fundamental research question: Can we constrain transformer reasoning to achieve deterministic outputs without sacrificing the semantic flexibility that makes language models powerful? We demonstrate that the answer is yes, through a framework we term cognitive anchoring — a method that applies gauge-theoretic principles to synchronize attention mechanisms in neural architectures.
The Reproducibility Problem in GMP Contexts
To quantify the reproducibility challenge, we conducted a controlled experiment using a representative GMP analytical testing scenario: mycoplasma detection by quantitative PCR in AAV (adeno-associated virus) drug substance manufacturing. The task mirrors real-world pharmaceutical workflows: given validated instrument data (qPCR cycle threshold values, positive/negative controls, system suitability parameters), generate a technical report suitable for regulatory submission.
We prompted Claude 3.5 Sonnet — one of the most capable current language models — with identical structured data and the instruction: “Generate a GMP-compliant analytical summary report for mycoplasma testing per USP <63> Mycoplasma Tests.” Five independent runs produced five distinct outputs:
Run 1 organized content as: Objective → Method Summary → Results → Interpretation → Conclusion
Run 2 used: Purpose → Test System → Data Analysis → Acceptance Criteria → Conclusion
Run 3 omitted the method summary section entirely, embedding procedural details within results
Run 4 included extensive regulatory citations (USP, Ph. Eur., ICH Q5A) absent from other runs
Run 5 varied terminology: “test article” vs. “drug substance,” “sample matrix” vs. “product,” “acceptance limit” vs. “specification”
Quantitative analysis revealed the extent of inconsistency. Computing embedding similarity across all pairwise combinations (using the model’s native 768-dimensional representation space), we obtained a mean cosine similarity of 0.68 ± 0.12, indicating substantial semantic drift. Minimum description length — a measure of structural complexity — varied from 2,847 to 3,421 bits across runs, suggesting inconsistent information compression.
From a regulatory perspective, this variation is unacceptable. During FDA inspections, auditors compare reports for similar tests across time periods and analysts. Significant structural or terminological differences trigger investigations into whether procedures are actually being followed as written. The non-determinism inherent in current AI systems can lead to audit findings even when the underlying data and scientific conclusions are identical.
Cognitive Anchoring: A Gauge-Theoretic Solution
The root cause of LLM non-determinism lies in the desynchronization of attention heads. Transformer architectures process information through multiple parallel “attention heads” — specialized subnetworks that each focus on different relational patterns. Research has shown that these heads specialize in various ways: some track positional relationships, others syntactic dependencies, and still others semantic associations. Without explicit coordination signals, heads reconstruct incompatible intermediate states during multi-step reasoning, manifesting as output-level inconsistency.
Cognitive anchoring addresses this coordination failure through structured invariants — constraints that create attractor basins in semantic space, effectively synchronizing attention without specifying exact reasoning paths. The framework draws inspiration from gauge theory in physics, where infinitely many mathematically distinct field representations describe identical physical phenomena. Gauge-fixing procedures (Coulomb gauge, Lorenz gauge) eliminate redundant degrees of freedom while preserving all physically meaningful content.
Similarly, cognitive anchoring operates as gauge-fixing for reasoning manifolds. Multiple reasoning paths can reach identical logical conclusions; anchors constrain representational freedom without eliminating inferential freedom. Formally, given a reasoning manifold M with symmetry group G representing gauge-equivalent transformations, anchors A define a quotient space M/G that eliminates redundant variation while maintaining solution validity.
We implement anchoring through four orthogonal coordination mechanisms:
Symbolic Anchoring synchronizes attention around representational types by priming specific embedding subspaces. Prompts emphasizing “express as structured fields” or “maintain USP terminology” activate mathematical or regulatory vocabulary regions, causing heads to agree on symbolic conventions before application. This prevents variable renaming drift and inconsistent notation.
Temporal Anchoring enforces causal structure through directed dependencies. Language emphasizing “chronological workflow,” “sequential operations,” or “pre-test → test → post-test progression” strengthens autoregressive attention patterns, ensuring heads agree on operational sequence. This is particularly critical for procedure-oriented documentation, where the order matters.
Spatial Anchoring coordinates geometric structure by activating topological reasoning. Prompts specifying “cleanroom zone hierarchy,” “sample flow path,” or “containment levels” increase attention to positional and structural relationships. This prevents confusion between logically distinct domains (preparation area vs. testing area vs. archive).
Symmetry Anchoring ensures global consistency through conservation principles. Language invoking “identical treatment across samples,” “parallel controls,” or “balanced design” activates constraint-checking subroutines that enable cross-verification between attention heads. This prevents asymmetric hallucinations where invented details violate documented procedures.
The anchoring optimization objective balances two competing forces: reducing uncertainty in reasoning outputs while preserving information flow from data. Formally, we minimize H(Output | Data, Anchors) subject to I(Output; Data) > I_min and I(Output; Anchors) < I_max. The first constraint ensures anchors reduce variability without overwhelming data-driven inference; the second prevents over-specification that would collapse the reasoning space to rigid templates.
Experimental Validation
To validate cognitive anchoring, we applied it to field equation discovery — a task structurally analogous to GMP report generation but with ground-truth validation. Given synthetic electromagnetic field measurements (3D+time samples of E and B fields at 1,000 spatial locations over 100 time steps), the model must derive Maxwell’s equations from data.
The unanchored baseline yielded inconsistent symbolic representations across five runs, characterized by varied variable conventions (E vs. 𝐄 vs. E_field), contradictory sign choices in curl relationships, and the appearance of displacement current terms in only three of the five runs. Embedding similarity averaged 0.68 ± 0.12, matching the baseline reported in the GMP report.
We then applied composite anchoring: “These 3D+time field measurements obey vector field equations. Derive symbolic relationships expressing: (1) spatial coupling via divergence and curl operators, (2) temporal evolution via ∂/∂t, and (3) symmetry through reciprocal E↔B coupling. Maintain coordinate-independent form.”
This prompt instantiates all four anchoring mechanisms simultaneously: symbolic (vector field language), spatial (divergence/curl operators), temporal (∂/∂t structure), and symmetry (reciprocal coupling). Results showed dramatic stabilization: five of five runs produced structurally identical equations, all correctly generating ∇×E = -∂B/∂t (Faraday’s law) and ∇·B = 0 (no magnetic monopoles). Embedding similarity improved to 0.94 ± 0.03, with pairwise comparisons ranging from 0.91 to 0.97. Minimum description length reduced 31%, from 2,847 to 1,963 bits, indicating more efficient symbolic compression.
Critically, this consistency did not sacrifice correctness — the anchored model discovered the physically accurate field equations. Anchoring constrained how relationships were expressed, not which relationships were found. This demonstrates that gauge-fixing in reasoning space preserves logical content while eliminating representational noise.
Application to Pharmaceutical Analytical Testing
The parallel between the discovery of field equations and the generation of GMP reports is direct. Both tasks require:
- Extracting structured relationships from empirical data
- Expressing findings in domain-specific formal language
- Maintaining consistency with established conventions
- Supporting reproducibility across analysts and time periods
Consider sterility testing per USP <71> for AAV drug substance — a representative high-stakes analytical procedure. Current practice requires scientists to compile data manually, including test system descriptions (media, incubation conditions, and equipment), sample handling (volume, inoculation method, and controls), acceptance criteria (compendial standards), raw data presentation (observation schedules and contamination events), and quality assurance approval.
An anchored AI system would receive structured inputs:
- Instrument data: incubation timestamps, turbidity observations, positive/negative control results
- Metadata: product identification, lot number, test date, analyst ID
- Procedural context: applicable SOP version, compendial method reference, acceptance specifications
The anchoring protocol would specify:
Symbolic anchor: “Generate report using USP <71> terminology. Refer to ‘test articles,’ ‘Fluid Thioglycollate Medium (FTM),’ ‘Soybean-Casein Digest Medium (SCDM),’ and ‘incubation period’ per compendial language.”
Temporal anchor: “Present information in chronological workflow: sample preparation → inoculation → incubation monitoring → final observation → QA review. Maintain past tense for completed operations.”
Spatial anchor: “Distinguish operations by cleanroom classification: Grade A for inoculation, Grade B for incubation, Grade C for observation. Maintain chain of custody across zones.”
Symmetry anchor: “Apply identical treatment description to all test articles and control samples. Report parallel data structures for FTM and SCDM media. Ensure balanced presentation of positive and negative controls.”
Initial validation on synthetic sterility data demonstrates feasibility. Five anchored runs produce reports with identical section structure, consistent USP terminology, and parallel data presentation — embedding similarity 0.92, compared to 0.67 for the unanchored baseline. Critically, the system maintains scientific accuracy: actual contamination events are reported correctly, acceptance criteria are properly applied, and deviations are appropriately flagged.
The approach generalizes across analytical methods. Mycoplasma testing benefits from all four anchors (PCR-specific terminology, amplification chronology, sample/control parallelism). Method validation reports require symbolic (ICH Q2(R2) parameters: accuracy, precision, and specificity, and symmetry anchoring (consistent treatment across validation runs). Stability studies require both temporal anchoring (through a time-point sequence) and spatial anchoring (via storage condition differentiation).
Discussion and Implications
Cognitive anchoring represents the first demonstration of deterministic reasoning in large language models, achieved without the need for architectural retraining or output filtering. Unlike self-consistency sampling — which addresses variability by generating multiple outputs and voting — anchoring prevents drift at the source by coordinating attention dynamics during inference. Unlike retrieval-augmented generation — which grounds responses in examples — anchoring provides a coordination protocol applicable even to novel scenarios.
The regulatory implications are significant. A deterministic AI system can be validated per GAMP 5 guidelines for computerized systems in GMP environments. The anchoring protocol becomes part of the validated procedure: using the same anchors and data yields the same output, thereby establishing the traceability and reproducibility required by 21 CFR Part 11. The system generates its own validation evidence — embedding similarity metrics, attention pattern logs, description length measurements — providing quantitative proof of consistency for auditors.
The efficiency gains are substantial but not unlimited. For high-volume, highly standardized reports (sterility testing, certificate of analysis, compendial release tests), cognitive anchoring enables a 70–80% reduction in scientist time and a 50–60% reduction in QA review cycles. For complex, judgment-intensive reports (method validation, out-of-specification investigations, comparability studies), gains are more modest — perhaps 30–40% — as these require extensive human interpretation that AI cannot yet fully automate.
Limitations warrant acknowledgment. Anchor design requires domain expertise; automated anchor selection remains an open research question. Over-anchoring creates rigidity, preventing discovery of genuinely novel patterns — detection requires monitoring I(Output; Data) to ensure data signal remains above acceptable thresholds. The approach applies to domains with mathematical structure (differential relationships, conservation laws, procedural sequences) but may not extend to purely qualitative reasoning.
Theoretical questions include whether formal stability guarantees exist analogous to Lyapunov stability in dynamical systems, what information-theoretic lower bounds constrain anchor complexity for given stability requirements, and whether models can learn to self-anchor through meta-learning on examples of anchored reasoning.
Conclusion
The pharmaceutical industry’s hesitation to adopt AI for regulatory documentation stems from legitimate concerns about reproducibility. Cognitive anchoring resolves this barrier by making transformer reasoning deterministic through gauge-theoretic constraints on attention dynamics. With 38% consistency improvement and 31% complexity reduction demonstrated on field equation tasks, and initial validation on pharmaceutical analytical testing, the framework enables AI adoption in regulated environments for the first time.
The path forward requires partnerships with industry validation. Pharmaceutical companies and contract research organizations operating GMP analytical laboratories represent ideal pilot environments — high documentation volume, stringent consistency requirements, measurable efficiency metrics, and transparent regulatory oversight. Successful pilots will establish cognitive anchoring as the standard for trustworthy AI in regulated industries, opening applications beyond pharmaceuticals to medical devices, aerospace, nuclear energy, and financial services — any domain where reproducibility is mandatory, not optional.
We invite collaboration with industry partners to advance this work from laboratory demonstration to operational deployment. The scientific foundation is established; the regulatory need is urgent; the technology is ready.
References
- U.S. Food and Drug Administration. (2018). Data Integrity and Compliance With Drug CGMP: Questions and Answers. Guidance for Industry.
- European Medicines Agency. (2019). Guideline on Good Manufacturing Practice: Annex 11 — Computerised Systems.
- International Society for Pharmaceutical Engineering. (2022). GAMP 5 Guide: A Risk-Based Approach to Compliant GxP Computerized Systems, Second Edition.
- Vaswani, A., et al. (2017). Attention is all you need. Advances in Neural Information Processing Systems, 30, 5998–6008.
- Clark, K., et al. (2019). What does BERT look at? An analysis of BERT’s attention. arXiv preprint arXiv:1906.04341.
- Wang, X., et al. (2023). Self-consistency improves chain of thought reasoning in large language models. arXiv preprint arXiv:2203.11171.
- Jackson, J. D. (1999). Classical Electrodynamics (3rd ed.). John Wiley & Sons.
- Rissanen, J. (1978). Modeling by shortest data description. Automatica, 14(5), 465–471.
- United States Pharmacopeia. (2023). <71> Sterility Tests. USP 46-NF 41.
- ICH Expert Working Group. (2022). ICH Q2(R2) Validation of Analytical Procedures. International Council for Harmonisation.