When Drugs Get Smart and Adaptive

I remember the first time I heard the term “smart drug” applied to insulin – the idea that a modified insulin could automatically adjust its activity based on a diabetic person’s blood sugar. As someone fascinated by new frontiers of biology, I was immediately hooked. Could we really make a drug that acts like a tiny feedback-controlled system inside the body? Over the past couple of years, this sci-fi concept has edged closer to reality. In this post, I want to share my excitement about the rise of self-regulating therapeutics enabled by programmable biology. I’ll walk through two in-depth examples that I find particularly groundbreaking:

  1. A glucose-sensitive insulin (codenamed NNC2215) that changes its shape depending on blood glucose levels – effectively a molecular dimmer switch for insulin activity. This insulin was described in an October 2024 Nature paper, and it offers a form of autonomous, closed-loop glycemic control in the body.

  2. Strand Therapeutics’ programmable mRNA therapy that behaves like an internal diagnostic-and-drug system. It uses logic gates encoded in mRNA to sense if a cell is cancerous (via its microRNA profile) and then conditionally expresses a powerful cytokine (IL-12) only in tumor cells, leaving healthy cells alone. This is basically genetic circuitry inside a drug, representing a new class of dynamic, cell-type-specific therapeutics.

Along the way, I’ll also discuss the broader principles behind these advances – from gene circuits and bioswitches that perform biological computation, to CRISPR-based logic and protease-activated drugs. We’ll see how these differ from simpler “smart delivery” systems and why embedding feedback control into therapeutics is such a big deal. Finally, I’ll touch on the challenges that remain (delivery, safety, predictability, manufacturing) and what this trend could mean for the future of medicine – a shift from static one-size-fits-all interventions to adaptive, responsive therapies.

Let’s dive in, starting with the glucose-sensitive insulin that I like to think of as a little molecular machine regulating itself in real time.

A Glucose-Sensitive Insulin “Switch”

One of the longest-running goals in diabetes research has been to create an insulin that turns itself on and off according to blood sugar levels. The problem it aims to solve is straightforward: if a diabetic patient injects too much insulin for their current needs, their blood glucose can plummet to dangerously low levels (hypoglycemia). Conventional insulin therapy is a balancing act – patients must constantly monitor glucose and adjust doses to avoid highs and lows, often erring on the side of caution to prevent nighttime hypoglycemia. Wouldn’t it be nice if insulin could sense when glucose is high (after a meal) and activate itself, but then automatically dial back when glucose falls, preventing an overdose?

That’s exactly what NNC2215 attempts to do. In the October 2024 Nature study, researchers from Novo Nordisk and collaborators engineered insulin with a built-in glucose-responsive switch. The design is elegant: they attached two extra pieces to the insulin molecule – a glucose-binding macrocycle and a glucoside (a glucose-like molecule tethered to insulin). Together these form a reversible “lock-and-key” mechanism that responds to ambient glucose.

Put simply, at low blood sugar, the insulin is held in an inactive “closed” state; at high blood sugar, it opens up into an active state and can signal cells to absorb glucose (Would love to put an image here but Squarespace wouldn’t allow me - another incentive to start a Substack soon).

The macrocycle is a kind of synthetic molecular ring (developed by chemists over decades to selectively bind glucose). In NNC2215, this macrocycle is chemically attached at a specific site on the insulin (amino acid B29, one of insulin’s tail ends). The insulin is also modified at another site (B1, the very start of the B-chain) with a glucoside – essentially an insulin-tethered sugar that can fit into the macrocycle like a key into a lock.

Here’s the clever bit: The macrocycle prefers to bind glucose, but in the absence of free glucose it will bind the tethered glucoside instead. So when your blood sugar is low, the macrocycle and the glucoside latch onto each other intra-molecularly, causing the insulin to fold in a way that blocks its receptor-binding surface. (In technical terms, the closed state causes a steric clash with the insulin receptor’s α-CT segment, which is crucial for insulin’s binding.) In this state, the insulin’s affinity for the insulin receptor is greatly reduced – it’s like the insulin is “off”.

But when glucose levels rise (for example, after a meal), glucose molecules in the bloodstream can enter the macrocycle’s pocket and displace the glucoside. Now the macrocycle is occupied by a free glucose, and the tethered glucoside is pushed out. The insulin molecule relaxes into an open conformation where its receptor-binding domains are exposed. In this high-glucose scenario, NNC2215’s affinity for the insulin receptor shoots up – it essentially turns “on” in response to the sugar signal. One scientist described it as not a binary light switch but a “dimmer” that smoothly ramps insulin activity up or down with glucose levels. The activity isn’t simply on/off; it’s proportional to how much glucose is around, much like how our pancreas’s beta cells would naturally respond (albeit via a different mechanism).

Structure-Function and Switch Performance

From a structure-function perspective, what’s remarkable is that the researchers were able to attach these bulky groups to insulin and still have it fold and function. Insulin is a small protein, and its ability to bind the insulin receptor depends on a precise conformation. By attaching the macrocycle at B29 and the glucoside at B1 (connected by short linkers), they effectively created a hinge or lid that covers the insulin’s binding interface in the closed state. The open vs. closed 3D models illustrate this: in the closed form, the macrocycle and glucoside snugly bind each other and sit in the way of the receptor. In the open form, the macrocycle has a glucose bound and the tethered glucoside is dangling out, leaving the insulin free to dock with the receptor.

How well does it actually work? In in vitro tests, NNC2215 showed about a 3.2-fold increase in insulin receptor binding affinity when glucose concentration was raised from 3 mM (low) to 20 mM (high). In fact, from 0 to 20 mM glucose the affinity change was even larger (~12.5-fold). This means the insulin’s potency is significantly higher in a high-glucose environment, which is exactly what we want. (By contrast, normal insulin has the same affinity regardless of glucose – it would happily keep driving glucose into cells even when blood sugar is already low.)

Crucially, this switching happens in a physiologically relevant glucose range (about 3–20 mM, roughly 54–360 mg/dL). That range covers normal fasting glucose up to well above typical post-meal levels in diabetes. One expert, Richard DiMarchi, noted that achieving such glucose sensitivity in this narrow window is “a benchmark in the field of macromolecular medicinal chemistry.” It’s worth highlighting that NNC2215’s response is reversible and repeatable – unlike glucose-triggered insulin pumps or polymer depots that release insulin in one big push, this modified insulin can continuously toggle between less-active and more-active forms as glucose levels ebb and flow.

In Vivo results: toward closed-loop glycemic control

In laboratory animals, the glucose-sensitive behavior of NNC2215 translated into real functional benefits. Experiments in diabetic rats and pigs showed that NNC2215 could lower high blood glucose effectively, but when the animals’ glucose started dropping toward normal, the insulin’s activity tapered off, avoiding hypoglycemia. In one comparative test, pigs were infused with glucose (to simulate feeding) and then given either NNC2215 or a conventional long-acting insulin (insulin degludec). When the glucose infusion was stopped, the degludec drove glucose below 3 mM (into hypoglycemic territory), whereas NNC2215 self-adjusted and stabilized the pigs around ~4.5 mM. Essentially, NNC2215 kept the animals’ sugars in a safe range without external intervention, whereas the normal insulin overshot and caused an unsafe dip.

This is a huge deal – it demonstrates autonomous, closed-loop glycemic control by the drug itself. Instead of a pump + sensor + algorithm, the molecule alone carries the feedback mechanism. The Nature paper reported that NNC2215 protected animals from hypoglycemia while still adequately reducing hyperglycemia during glucose tolerance tests. In diabetic rats, it blunted glucose “excursions” (spikes) and then backed off as levels normalized.

Seeing those results felt almost like witnessing a pancreas in action – but one made of chemistry and protein engineering. It’s not a perfect mimicry of endogenous insulin secretion (NNC2215’s switch is more gradual, and it has a certain baseline activity), but it’s a big step closer to the dream of a “set-and-forget” insulin shot that manages itself.

Of course, there are caveats. Speed is one – NNC2215 still has to circulate and bind/unbind, so there might be some lag in response (though likely faster than polymer-based glucose-responsive systems, which rely on glucose diffusing into a depot). Another is strength of response – a 3-fold change in activity might not be enough on its own to perfectly control diabetes in all situations. However, even a partial self-regulation can allow patients to safely take more insulin upfront (covering meals better) without constant fear of a crash. One can imagine pairing NNC2215 (which partly “auto-titrates” basal insulin) with fast-acting meal boluses, or using it in pumps for added safety.

Finally, the researchers acknowledge that we need to see if NNC2215 works as a chronic therapy. Will it remain stable and non-immunogenic in humans over many injections? The molecule is more complex than human insulin, so there’s potential for the immune system to see it as foreign. Additionally, manufacturing such a conjugate at scale is non-trivial – the macrocycle’s synthesis was initially very difficult, though the team optimized it to 95% yield with clever chemistry. These practical challenges (which I’ll revisit later) mean NNC2215 isn’t hitting the pharmacy tomorrow. But conceptually, it has proven that a self-regulating protein therapeutic is possible, ending decades of “smart insulin” research with little to show. As one news headline aptly put it, this “‘smart’ insulin prevents diabetic highs – and deadly lows.”

With that in mind, let’s turn to the second example – which, if anything, is even more like science fiction: an mRNA that computes whether it’s in a cancer cell or not, and then makes a drug only if the answer is “yes.”

Strand Therapeutics’ IL-12 Logic Circuit

If the glucose-responsive insulin is a molecular dimmer switch, Strand Therapeutics’ approach is like embedding a biological computer program in a therapy. The concept blew my mind when I first read about it: take a single mRNA (the same kind of messenger RNA used in vaccines), and encode within it a logic circuit that controls whether the mRNA produces a protein or self-destructs – depending on what type of cell it’s in. The goal is to create a conditional cancer immunotherapy that turns itself on only in tumor cells and stays off elsewhere, thereby maximizing impact on the cancer while minimizing toxicity.

The challenge: IL-12 is potent but dangerous

Strand’s flagship programmable mRNA (often referred to as STX-001 or STX-003 in various reports) encodes Interleukin-12 (IL-12), a powerful immune-stimulating protein. IL-12 is known to activate killer T cells and NK cells and can trigger strong anti-tumor responses. However, IL-12 is so potent that if you inject it systemically, patients get very sick – severe inflammation, organ damage, etc. (In fact, high-dose IL-12 in the bloodstream can cause life-threatening toxicity, which halted earlier IL-12 therapies.) The key to using IL-12 against cancer is localization: you want IL-12 only in the tumor microenvironment, not coursing through the body. Some approaches to achieve this include injecting IL-12 directly into tumors or making “pro-IL-12” molecules activated by tumor enzymes (more on that later). Strand’s idea is different: use a systemically delivered mRNA, but program that mRNA such that it only produces IL-12 inside cancer cells and not in normal cells like those in the spleen or liver.

How can an mRNA know whether it’s in a cancer cell or a normal cell? It turns out that different cell types (and healthy vs. cancerous cells) have distinct patterns of small regulatory RNAs called microRNAs. These ~22-nucleotide RNA snippets are like fingerprints of a cell’s identity – for example, a certain microRNA might be abundant in healthy liver but nearly absent in a liver tumor, or vice versa. Strand’s team exploits this by encoding microRNA sensor sequences into the therapeutic mRNA itself. Essentially, the mRNA can “read” the cell’s microRNA levels and then decide whether to express IL-12 or to shut itself down.

Logic gates in mRNA

The mRNA logic circuit functions on a simple principle: if the cell is an unwanted target (e.g., a healthy cell in the spleen), the circuit prevents IL-12 expression or even destroys the mRNA; if the cell is the intended target (a tumor cell), the mRNA remains intact and produces IL-12. Strand describes it as the mRNA “sensing the environment” – if it senses the right signature, it “follows its programming” to make the drug; if not, it “cleaves itself and is degraded”. In practice, this is done by including microRNA binding sites (also known as “microRNA response elements”) in the mRNA’s sequence. These can be placed in regions like the 3′ untranslated region of the mRNA. The idea is:

  • In cells where a particular microRNA is present at high levels, that microRNA will bind to the complementary sites on the therapeutic mRNA. This triggers the cell’s natural RNA interference machinery (RISC complex) to slice up or repress the mRNA, preventing IL-12 protein from being made. Essentially, the mRNA self-destructs in those cells.

  • In cells where that microRNA is low or absent, it cannot bind the mRNA, so the mRNA remains intact and gets translated into IL-12 protein.

By using multiple microRNA sensors in combination, one can create more complex logic. For instance, the mRNA might have target sites for MicroRNA A and MicroRNA B, both of which are abundant in healthy tissues but scarce in tumors – so only in cells where neither A nor B is around (i.e. likely a tumor cell) will the mRNA survive to produce IL-12. This effectively functions as an AND gate on the absence of those microRNAs (or conversely an OR gate for self-destruction in any cell that expresses any of those markers). According to Strand’s CEO, they computationally design circuits that respond to “a certain number of different miRNAs that are either over or under expressed” as markers of a given cell type. It’s like the mRNA has a multi-factor checklist: “Am I in a cell with miR-X low AND miR-Y low AND miR-Z high? If yes (this looks like a cancer cell), proceed to make IL-12. If not, abort!”

Importantly, Strand’s mRNA drugs use a self-replicating mRNA platform (derived from RNA viruses). This means each delivered mRNA can amplify itself inside the cell (like how certain viral RNA genomes replicate), yielding more copies that produce IL-12. Combining replication with logic gating is quite sophisticated – it ensures that in target cells you get a strong, sustained IL-12 production (amplification), but in off-target cells the replication is shut down by the microRNA triggers. It’s as if the mRNA carries code to “check what cell I’m in; if it’s wrong, commit suicide; if it’s right, copy myself and deploy the weapon (IL-12).” And all of this is happening at the RNA level, without needing any external control. How cool is that?

Results: Hitting Tumors, Sparing Healthy Tissue

The true test of this system is in animal studies: does it actually localize the IL-12 effect to tumors and reduce systemic toxicity? The answer, so far, is very encouraging. In mouse tumor models, a “circuited” IL-12 mRNA (with the microRNA logic) was compared to an “uncircuited” version (the same mRNA but without the special sensor elements). Both were delivered intravenously in lipid nanoparticles (LNPs) that largely accumulate in liver, spleen, and tumors. Here’s what they saw:

  • Tumor targeting: The circuited mRNA expressed IL-12 just as effectively in tumors as the uncircuited one. In melanoma-bearing mice, both treatments led to similar IL-12 levels in the tumor and equally strong tumor growth control. In other words, the added genetic circuit did not diminish the anti-tumor punch – a critical point for efficacy.

  • Off-target repression: In the spleen (a frequent site of immune toxicity), the circuited mRNA produced 40+ times less IL-12 protein than the normal mRNA. The circuit drastically detargeted the spleen, ensuring IL-12 levels there remained low. Despite that, in the tumors, IL-12 was still high – meaning the ratio of tumor-to-spleen IL-12 improved hugely.

  • Safety signals: In normal (non-tumor-bearing) mice and even in cynomolgus monkeys, the circuited IL-12 mRNA yielded dramatically less systemic cytokine exposure. Circulated IL-12 in blood was ~99% lower with the circuit vs. the unmodified mRNA, and downstream inflammatory signals like IFN-γ were ~95% lower. Essentially, the engineered mRNA kept the IL-12 protein localized and out of the circulation. In mice, this translated to much better tolerability (less sickness behavior, etc.). In monkeys, it’s a strong indication that this approach could be safe at doses that would have been very toxic otherwise.

  • In vitro confirmation: When they transfected cancer cells (like A549 lung cancer cells or patient-derived tumor organoids) with circuited vs. uncircuited mRNA, both produced robust IL-12. But in cells from normal tissue that express the microRNA, the circuit would repress expression. This shows the logic works as intended – “express payload in cancer cells, repress in healthy cells.”

Together, these results suggest that the mRNA circuit can “reprogram the tumor microenvironment” with IL-12 without causing systemic cytokine exposure. In mouse tumor models, this leads to strong anti-tumor efficacy (especially when combined with checkpoint inhibitors) and minimal side effects. In fact, Strand has reported that their IL-12 mRNA therapy, when injected into tumors or given IV with circuits, induced immunogenic tumor cell death and recruitment of T cells, boosting the effect of PD-1 checkpoint blockade. It’s basically acting like a localized immunotherapy “grenade”, going off only in the tumor.

Strand’s approach is now moving through clinical trials. They announced the first-in-human Phase 1 data recently, using an IL-12 mRNA (called STX-001 when given intratumorally, and STX-003 for IV with circuits). The interim results in patients will tell us if human tumors respond similarly and – critically – whether side effects are indeed blunted. The promise is huge: IL-12 is known to be effective at rallying the immune system against cancer, but it’s like a fuse that burns too hot. By adding a smart circuit, we might finally be able to deploy IL-12 systemically in a safe way. As Strand’s team put it, this could “enable precise control of the location, timing, and intensity” of a potent therapeutic inside the patient. Essentially the drug has a built-in GPS and logic board – it knows where it is and makes decisions accordingly.

I find this especially exciting because it represents a paradigm shift: instead of engineering the patient (as in CAR-T cell therapies or oncolytic viruses), we are engineering the drug itself with conditional behavior. It’s a drug that contains an if-then statement! The mRNA is like, “if cancer, then IL-12; else, do nothing.” And it’s doing this using the cell’s native biomolecules (microRNAs) as inputs – harnessing nature’s own regulatory code. This opens the door to a whole new class of “smart therapeutics” where specificity is not just achieved by targeting an address (like an antibody homing to a receptor) but by actual logical computation inside the body.

Beyond these examples: bioswitches and biological circuits as therapeutic tools

The glucose-responsive insulin and the programmable IL-12 mRNA are two leading examples, but they are part of a broader movement in biotech to create adaptive therapies. The common theme is integrating a sensor + effector + logic all in one therapeutic system. Let’s briefly survey some other approaches and principles emerging in this space:

  • Gene Circuits in Cell Therapies: Synthetic biology has given us the ability to implant genetic circuits into living cells (like engineered T cells or stem cells). For example, researchers have built CAR-T cells with logic gates – requiring two antigens to activate (AND gate) or having a NOT gate to avoid attacking normal cells that have a certain marker. There are CAR T designs that only kill cells if they detect combo A and B, or that kill and include a self-destruct if conditions aren’t right. These are not drugs in a vial, but living cells engineered with internal decision-making circuits. They highlight the same goal: safer, more precise action by requiring multiple signals (antigen A and low oxygen and inflammatory signal B, etc.) for full activation. Some of these circuits use synthetic promoters, transcription factors, and even CRISPR-based regulators to implement logic.

  • CRISPR-based Transcriptional Logic: A lot of excitement surrounds using CRISPR/Cas systems not just to edit genes but to control gene expression in a programmable way. By using a catalytically “dead” Cas9 (dCas9) fused to activator or repressor domains, scientists can target any gene’s promoter and turn it on or off. Stringing multiple gRNAs together or designing conditional gRNAs yields logical control over gene networks. In essence, CRISPR gives us a plug-and-play toolbox to build gene circuits. As a 2020 review put it, this is paving the way to a “new class of transcriptional therapeutics” – the dream of pinpoint control over gene expression in vivo is becoming reality. For instance, one could envision a gene therapy where a dCas9 circuit will only activate a tumor suppressor gene if it detects a cancer-specific microRNA and a hypoxic environment – an internal multi-sensor logic. While much of this is still in research phases, it’s only a matter of time before CRISPR-driven circuits enter therapeutic trials (some safety switches using CRISPR-based kill-switches in cell therapies are already being tested).

  • Small-Molecule-Controlled Proteins: Another strategy for “smart” therapeutics is designing proteins that can be toggled or guided by a benign small molecule. One example is drug-inducible switches for CAR-T cells – e.g., splitting a CAR into two halves that only come together (and become active) when a small molecule dimerizer is present. The clinician can then administer the small molecule to turn the CAR-T on, and withdraw it to turn them off, achieving a feedback-like control externally. Similarly, there are allosteric switches engineered into enzymes or signaling proteins that make them active only when a specific small molecule ligand (which by itself is inert) binds. These systems aren’t autonomous (since you still add the drug to control them), but they add a layer of control that can be tunable. A patient could have an implant or pump releasing the small molecule in response to their vitals, effectively creating a closed-loop. We’ve also seen PROTACs (proteolysis-targeting chimeras) where a small molecule causes disease proteins to be tagged for destruction – a sort of conditional effect only when the drug is present. The general idea is to move from drugs that irreversibly do one thing, to drugs that can be modulated in time or intensity by another agent.

  • Protease-Activated Biologics: I hinted earlier at “pro-IL-12” approaches. Companies like Xilio and CytomX are developing protease-activated cytokines and antibodies. These are large protein drugs that are fused to an inhibitory domain (a “mask”) via a linker that can be cut by enzymes abundant in the tumor microenvironment (like matrix metalloproteinases). In healthy tissues, the drug stays masked and inactive; when it drifts into a tumor, tumor-specific proteases cleave the linker, removing the mask and unleashing the drug’s activity. For example, a Pro-IL-12 has IL-12 fused to part of an antibody that blocks its receptor-binding site; an MMP in the tumor chops off the blocking part, activating IL-12 right there. These are essentially physical logic gates – they execute an IF “tumor protease present” THEN “activate drug” operation. Early data on a tumor-activated IL-12 (e.g., XTX-301) show it can indeed widen the therapeutic window, hitting tumors hard while sparing normal tissue. Similarly, masked antibodies (Probodies) stay inert until proteases in the tumor unmask the binding site, reducing off-tumor toxicity. These are simpler than gene circuits, but they share the same principle: conditional activation based on the disease environment.

  • Gene and RNA “Computers”: Beyond microRNA sensors like Strand’s, other nucleic-acid based logic circuits have been demonstrated in lab models. For instance, DNA or RNA strand-displacement circuits can be used as diagnostics or therapeutics that release outputs (like a therapeutic siRNA) only if certain combinations of RNA inputs (perhaps disease markers) are present. Synthetic mRNA-based riboswitches can sense metabolites or proteins and change gene expression accordingly. While not yet in humans, the field of molecular computing is advancing such that one day we might inject a nano-scale “computer” that makes therapeutic decisions cell by cell.

The key distinction in all these is between responsive delivery and true feedback control. A lot of so-called “smart drug delivery” systems have been devised – for example, nanoparticles that release drug faster at lower pH (to target acidic tumor tissue), or hydrogels that dissolve and dump insulin when glucose is high. Those are useful, but they are often one-time or open-loop responses. They don’t truly modulate dosage continuously up and down; they just release drug under certain conditions. The self-regulating systems we’ve been discussing, however, have an embedded feedback loop. NNC2215 continuously binds or releases glucose such that it equilibrates to the right amount of activity for the current glucose concentration. Strand’s mRNA continuously gets turned over and re-made; if a cancer cell starts expressing a certain microRNA, that input will immediately throttle the circuit. This is more akin to a thermostat (maintaining temperature by on/off feedback) rather than a bag of ice that just slowly melts when it’s hot. Both can cool a room initially, but the thermostat is smarter in the long run.

To illustrate: a “smart” glucose-responsive insulin depot might release a burst of insulin when glucose crosses a threshold, but once that insulin is released, it’s out – there’s no turning it back down until the next dose. NNC2215, in contrast, binds and releases insulin in a reversible way depending on glucose, acting continuously. This ability to go back and forth (on in high state, off in low state, repeatedly) is what makes it a true closed-loop system embedded in the molecule.

Challenges and Future Outlook

We are on the cusp of a new era of therapeutics where drugs are not just static chemical entities, but dynamic agents that sense, decide, and adapt. It’s an incredibly exciting frontier – but it comes with substantial challenges that scientists and engineers are actively working to overcome. In my view, some of the key hurdles and considerations include:

  • Delivery: All the fancy logic in the world is useless if you can’t deliver the therapeutic to the right place. For NNC2215, delivery is straightforward (it’s an injectable insulin, so subcutaneous injection), but for Strand’s mRNA and similar circuits, drug delivery technology is crucial. Strand uses lipid nanoparticles for mRNA delivery, which tend to accumulate in certain organs (liver, spleen) and also at tumor sites (due to leaky vessels in tumors). Ensuring that enough of the mRNA actually reaches most tumor cells (and not other off-target organs) is an ongoing challenge. Targeted delivery vehicles or local administration strategies might be needed for different indications. Delivery is also an issue for large engineered proteins or virus-like particles – can they penetrate tissue, cross biological barriers, and reach all disease sites? We might need innovative carriers (nanoparticles, cell-based delivery, etc.) to fully harness these adaptive therapies.

  • Safety and Immunogenicity: By adding complexity (foreign sequences, new chemical groups) to therapeutics, we also introduce new risks. The immune system might see a modified insulin or a replicating mRNA as an invader. For instance, mRNA therapies can trigger innate immunity (though companies modify the RNA and use suppressors to mitigate this). Proteins like NNC2215 could potentially lead to anti-drug antibodies that neutralize it or cause allergic reactions. Extensive safety testing is needed to ensure that the “self-regulation” mechanisms don’t misfire. What if NNC2215 doesn’t turn off fast enough – could it still cause late-onset hypoglycemia? What if Strand’s circuit in rare cases fails and expresses IL-12 in a normal organ – could that lead to serious toxicity? Redundancies (multiple safety switches) may be built in to prevent one failure from causing harm. Regulators will understandably be cautious – these are more complex than a pill, so demonstrating consistent behavior and safety will require robust data.

  • Predictability and Tuning: Biological systems are notoriously variable. Patient-to-patient differences (in microRNA levels, metabolic rates, etc.) might affect how these smart therapies perform. One diabetic’s glucose dynamics might differ from another’s; will NNC2215’s “3-fold affinity change” be enough for someone who has very brittle diabetes, or will it need tuning? Similarly, microRNA profiles vary between tumors – Strand’s circuit is presumably designed for an “average” tumor vs. normal signature, but a subset of patients might have atypical expression and thus the logic could mis-classify some cells. The tuning of thresholds is something we usually do with devices (adjust an insulin pump’s algorithm, for example); with a molecule, you have to get the tuning right in the design. Future iterations might include controllable parameters – e.g., a drug that you can administer a second agent to tweak its sensitivity up or down in a given patient. We might also see learning algorithms integrated, where data from initial doses inform adjustments to subsequent doses (blending biochemical feedback with software feedback).

  • Manufacturing Complexity: These advanced therapeutics are often much more complex to manufacture than traditional small-molecule drugs or biologics. NNC2215 requires multi-step organic synthesis to attach the macrocycle and glucoside in specific ways. It’s doable (they achieved 95% purity in the macrocycle synthesis finally), but scaling that to industrial levels cost-effectively could be challenging. Programmable mRNAs require high-quality in vitro transcription and LNP formulation – which, thanks to mRNA vaccines, is now feasible at large scale, but self-replicating RNAs are longer and may have additional QC considerations (ensuring no replicative competent virus forms, etc.). Cell therapies with gene circuits require cell manufacturing plus genetic engineering steps. Overall, the cost of goods for these smart therapies might be higher, at least initially, and manufacturing processes will need to be refined for consistency.

  • Regulatory and Ethical Aspects: We’re effectively giving drugs a “decision-making” capability. This blurs the line between a drug and a device (or even a synthetic organism). Regulatory agencies will have to consider new frameworks for approving such products. How do you classify an mRNA that’s also a logic circuit? It’s not a typical biologic. There may be concerns about containment (especially for replicating systems – though Strand’s replicon RNAs are not viruses and can’t spread between cells, which is good). Ethically, we always must consider unintended consequences: if a therapy is too autonomous, could it behave in ways we don’t anticipate? For example, a self-replicating mRNA – we must ensure it eventually stops (perhaps by design, replicons dilute out after a few cell divisions or contain a “kill switch” after X replications). Safeguards in design will be as important as the primary function.

Despite these challenges, the trend is undeniably toward more “adaptive, feedback-embedded systems” in medicine. We are essentially programming biology to treat itself. Instead of blasting a disease with a high dose and hoping we guessed right, we introduce a smart agent that modulates its activity as needed – potentially in real time within the patient. This could mean treatments that are both more effective and safer. We’ve already seen the power of feedback control in devices (insulin pumps with glucose sensors dramatically improve diabetes outcomes by minimizing human error). Bringing that concept inside the drug molecule or therapeutic cell is the next logical step.

In the future, I imagine a whole therapeutic ecosystem of “Bio-IP” (Biological Intelligence) drugs. For autoimmune diseases, maybe a therapeutic protein that turns itself off when it detects certain inflammatory signals (preventing over-suppression of the immune system). For neurodegenerative diseases, perhaps a gene therapy that senses neuronal stress and adjusts its output accordingly. We might even combine multiple circuits: a cancer therapy cell that not only targets the tumor but also signals if it’s overwhelmed, prompting a second wave of a different drug. These combinations of internal and external feedback loops could make therapies far more robust to the complexities of human disease.

To end on a personal note: as someone who has spent years reading about incremental drug improvements, seeing therapeutics that contain actual logic operations is both surreal and thrilling. It brings the programmability of software to the wet, messy world of biology. We often call cells “tiny machines”, but now we’re literally borrowing concepts from computer science to enhance medicine – writing if-else conditions in DNA/RNA, designing molecular switches and circuits. It feels like we’re entering an era where medicine is not just about chemistry or biology alone, but about information processing in the body. The rise of self-regulating therapeutics is a key part of that evolution.

We have a long way to go, but the successes of NNC2215 and Strand’s mRNA therapy show that this isn’t just a theoretical exercise – it works in living animals, and hopefully soon in patients. In the coming decade, as these therapies progress through clinical trials, we’ll find out how well the promise translates to patient outcomes. I, for one, am optimistic that we’re witnessing the beginning of a shift: a move from static interventions (where we give a dose and cross fingers) to interactive treatments that continually respond and adjust to the patient’s needs. It’s medicine with a built-in brain – and that could be transformative for diseases like diabetes, cancer, and beyond.

Next
Next

AlphaFold 3 and the New Era of AI-Powered Drug Discovery