DREADDs — designer receptors exclusively activated by designer drugs — have reshaped the landscape of neuroscience research. By enabling precise, reversible control over specific cell populations, these engineered GPCRs have accelerated discoveries in brain function, behavior, and neuropsychiatric disease. Over the past 15 years, DREADDs have evolved in selectivity, delivery mechanisms, and clinical potential. Yet questions about off-target effects, translational barriers, and long-term safety remain. This article traces the rise of DREADDs from Roth’s original designs to today's miniaturized, humanized, and peripheral-ready variants—charting both the breakthroughs and the work ahead to bring this chemogenetic innovation to the clinic.
What Are DREADDs and Why Do They Matter?
Designer receptors exclusively activated by designer drugs (DREADDs) are engineered G protein–coupled receptors (GPCRs) that have been modified to respond selectively to synthetic ligands rather than endogenous neurotransmitters. Most commonly derived from muscarinic receptors, these tools allow researchers to activate or inhibit specific cellular pathways with exceptional precision. Once expressed in targeted cells — typically via viral vectors — DREADDs can be modulated by systemically administered ligands that are otherwise inert in the organism, such as clozapine-N-oxide (CNO) or Compound 21 (C21), thereby enabling reversible control of cellular activity without affecting unrelated systems.1
DREADDs are part of a broader class of neuromodulatory technologies known as chemogenetics, which contrast with optogenetics in several important ways. While optogenetics uses light-sensitive ion channels and requires the implantation of fiber optics or LEDs, DREADDs achieve cell-specific activation or inhibition through a minimally invasive pharmacological approach. This provides several distinct advantages, particularly for in vivo studies in freely moving animals or potential clinical applications in humans.
Key benefits of the DREADD system include temporal control over signaling cascades (ranging from minutes to hours), spatial selectivity through promoter or vector targeting, noninvasive ligand administration, and the ability to engage intracellular signaling pathways beyond simple depolarization. Depending on the DREADD construct used — such as Gq-, Gi-, or Gs-coupled receptors — researchers can modulate neuronal excitability, synaptic transmission, or gene expression with remarkable specificity.
Since their introduction in the early 2000s, DREADDs have seen widespread uptake in the neuroscience community, with hundreds of publications applying them across diverse research areas, from basic circuit mapping to disease modeling. Their rising popularity reflects both their experimental flexibility and the growing interest in chemogenetics as a foundation for next-generation therapeutics.2,3
The Origins and Engineering of DREADDs
The DREADD platform was first conceptualized as a means to achieve precise, cell-type–specific modulation of GPCR signaling using ligands that would not otherwise interact with endogenous receptors. Initial development efforts focused on muscarinic receptor backbones, culminating in several now widely used constructs: hM3Dq, a Gq-coupled excitatory receptor; hM4Di, a Gi-coupled inhibitory receptor; and hM3Ds, a Gs-coupled receptor designed to stimulate cAMP production.1,4 Each of these variants was engineered to be unresponsive to endogenous acetylcholine but highly sensitive to synthetic agonists, allowing selective engagement of downstream signaling.
These receptors were designed through targeted mutations that reshaped the ligand-binding domain, enabling them to accommodate molecules such as clozapine-N-oxide while eliminating sensitivity to native neurotransmitters.5 The structural modifications retained the receptor’s ability to recruit intracellular effectors upon activation while rendering it functionally inert under physiological conditions. This innovation opened the door to highly controlled pharmacological interventions at the level of discrete cell populations.
The most common ligands used to activate DREADDs include CNO, its active metabolite clozapine, and the more recently developed C21. Each presents distinct pharmacological challenges. Although CNO was originally believed to be inert in mammals, it has since been shown to back-convert to clozapine in vivo, leading to off-target effects due to clozapine’s promiscuous receptor profile.6,7 C21, developed as a more selective alternative, offers improvements in pharmacokinetics and blood–brain barrier penetration, but recent studies have also highlighted its potential for non-specific effects in some contexts.
Once activated by ligand binding, DREADDs initiate intracellular signaling through their respective G protein pathways. Gq-coupled DREADDs like hM3Dq stimulate phospholipase C activity, leading to increased intracellular calcium and neuronal excitation. In contrast, Gi-coupled DREADDs, such as hM4Di, inhibit adenylyl cyclase, reduce cAMP levels, and often produce hyperpolarization via activation of G protein–gated inwardly rectifying potassium (GIRK) channels. Gs-coupled variants like hM3Ds promote cAMP accumulation, enabling a third mode of pathway-specific activation.8
One of the defining strengths of the DREADD system lies in its temporal flexibility. Ligands can be administered to induce acute or sustained changes in activity, depending on dosing and ligand choice, and the effects are typically reversible within hours. This combination of pharmacological control and molecular specificity continues to make DREADDs a valuable tool in both experimental and translational neuroscience.
Applications in Neuroscience Research
Since their introduction, DREADDs have been widely adopted in neuroscience research to unravel the function of specific neural circuits and their contributions to behavior and disease. The ability to selectively manipulate neuronal activity in vivo has made chemogenetics a transformative tool for both fundamental research and translational modeling.
One of the earliest and most influential applications of DREADDs has been in circuit mapping and behavioral studies. By enabling researchers to activate or silence discrete populations of neurons, DREADDs provide a means to establish causal links between neural activity and behavioral outcomes. This approach has been applied across a broad array of paradigms, including investigations of anxiety, fear conditioning, reward processing, feeding behavior, and sleep–wake regulation.6,9,10 These studies have helped define the roles of specific neuronal subtypes and projection pathways in complex behavioral phenomena, revealing mechanisms that were previously inaccessible using traditional pharmacological or lesion-based methods.
In addition to basic circuit analysis, DREADDs have emerged as powerful tools in disease modeling. In Parkinson’s disease, DREADDs have been used to selectively activate dopamine D1 receptor–expressing neurons, leading to improvements in motor function in preclinical models. Notably, the development of humanized Gs-coupled DREADDs has allowed for long-term therapeutic studies that simulate clinical treatment paradigms.11,12 Similarly, DREADD-based inhibition of hyperexcitable neuronal populations has shown promise in epilepsy models, with studies demonstrating reduced seizure frequency and severity following targeted Gi-coupled receptor activation.13
Beyond movement disorders and epilepsy, DREADDs have also been used to interrogate circuits implicated in depression, schizophrenia, and neurodevelopmental conditions. By modulating activity in limbic and cortical structures, researchers have identified network-level changes associated with affective symptoms and cognitive dysfunction.8,10 These studies underscore the versatility of chemogenetics in modeling psychiatric disease mechanisms and testing the effects of targeted interventions.
While most DREADD applications have focused on the central nervous system, recent innovations have begun to extend their use to peripheral tissues. The development of the hydroxycarboxylic acid receptor DREADD (HCAD) represents a major step forward in this direction. Based on the HCA2 receptor, which is expressed predominantly in the immune system and minimally in the brain, HCAD allows for selective modulation of immune and metabolic processes without unintended central effects.14 Other efforts are exploring DREADD-based control in organs such as the liver, heart, and gastrointestinal tract, opening new avenues for organ-specific neuromodulation.15
These emerging peripheral applications expand the relevance of DREADDs beyond neuroscience, suggesting potential roles in immunology, endocrinology, and cardiology. As delivery platforms improve and receptor variants are refined for different tissue contexts, the impact of chemogenetics may grow well beyond its neurological roots.
Evolving the Platform: Innovations and Technical Progress
As chemogenetic approaches continue to mature, researchers have introduced a range of refinements to the DREADD platform to expand its utility, improve delivery efficiency, and address limitations related to immunogenicity, size constraints, and specificity. These technical innovations are paving the way toward broader applications in both preclinical and clinical settings.
Recent efforts have focused on engineering more compact receptor constructs to overcome the size limitations of adeno-associated virus (AAV) vectors. Traditional DREADD systems, especially when combined with fluorescent reporters or regulatory elements, can exceed the packaging capacity of AAVs. To address this, new miniaturized DREADDs have been developed that retain functional efficacy while reducing the genetic payload. These streamlined designs allow both excitatory and inhibitory receptor variants to be delivered simultaneously using a single AAV vector, facilitating bidirectional control of neuronal activity within the same cell population or brain region.14
Another major advancement is the creation of fully humanized DREADDs, such as hM3Ds. These receptors are designed to minimize the risk of immune responses that could arise from the use of non-human or chimeric proteins, a key consideration as DREADD-based therapies move closer to clinical translation. The hM3Ds variant has demonstrated robust signaling capacity while maintaining low immunogenicity profiles, making it a promising candidate for therapeutic use in humans.11
Beyond these structural innovations, researchers are leveraging synthetic biology to generate libraries of DREADD variants with customized signaling outputs. These efforts include optimizing receptor-ligand binding, tuning downstream G protein coupling, and designing receptors with enhanced stability or altered trafficking properties.15,16 Such tailored receptors offer more precise modulation of specific signaling pathways, which could enable tissue- or disease-specific interventions with fewer off-target effects.
Parallel to receptor design, advances in delivery platforms have played a crucial role in expanding the reach and reliability of DREADDs. AAVs remain the most widely used vectors owing to their neuronal tropism, low immunogenicity, and compatibility with long-term expression. However, other viral systems, including lentivirus and herpes simplex virus (HSV), are also employed depending on the experimental or therapeutic context. Integration with Cre-lox systems has further enabled conditional expression of DREADDs, allowing researchers to target specific cell types, developmental stages, or brain regions.9,13
To support spatial and temporal precision, region-specific promoters and recombinase-dependent expression strategies are often used. These techniques ensure that DREADDs are only expressed in desired populations, reducing the risk of unintended modulation. Long-term expression, a critical factor for potential clinical use, has also shown promise. Studies in non-human primates have demonstrated stable DREADD expression and sustained functionality over extended periods without apparent toxicity or loss of efficacy.12
Together, these innovations represent a significant evolution in the DREADD platform — enhancing its precision, scalability, and safety while laying the groundwork for next-generation chemogenetic therapies.
Challenges and Controversies
Despite the remarkable versatility and utility of DREADDs in research, several challenges and controversies continue to shape discussions around their broader application, particularly in translational contexts. These issues fall into three major categories: ligand-related concerns, expression-related variability and toxicity, and barriers to clinical adoption.
One of the most persistent concerns relates to the pharmacological profile of the ligands used to activate DREADDs. Initially, CNO was believed to be pharmacologically inert in mammals, making it an ideal candidate for selective receptor activation. However, subsequent studies revealed that CNO is rapidly converted in vivo to clozapine, a compound with high affinity for numerous endogenous receptors, including serotonergic, dopaminergic, and muscarinic subtypes.1,4 This metabolic back-conversion undermines the assumption of pathway specificity and introduces potential confounds, especially in behavioral experiments. C21 was developed in part to address this issue, offering improved selectivity and brain penetration. Still, off-target effects have been reported for C21 as well, including its interaction with endogenous receptors that can produce physiological side effects.6,7,14 Notably, C21 has been shown to induce acute diuretic responses via interaction with vasopressin receptors in wild-type mice, further highlighting the need for caution when interpreting chemogenetic results.14
Another critical area of concern is variability in expression and potential toxicity related to DREADD delivery. While DREADDs are generally considered safe, long-term expression in animal models has revealed occasional immune responses or changes in cellular function that may complicate chronic use. Studies in non-human primates have begun to explore these risks in more clinically relevant systems, identifying issues such as localized inflammation and variability in receptor expression that could affect both efficacy and safety.11,12 These effects may depend on factors such as vector type, promoter strength, expression duration, and the specific tissue targeted. Additionally, inconsistency in transgene expression across individuals can lead to variable outcomes, posing a challenge for reproducibility in both research and potential clinical use.
Finally, significant translational hurdles remain before DREADDs can be widely adopted in therapeutic settings. As with any gene-modifying technology, DREADD-based interventions must navigate a complex regulatory environment, including oversight related to vector manufacturing, delivery protocols, and long-term safety monitoring. Compounding these challenges are species-specific differences in ligand pharmacokinetics, which can affect both drug efficacy and safety when transitioning from animal models to human applications.3 Moreover, designing clinical trials for DREADD-based therapies raises additional ethical and logistical questions, particularly around informed consent, vector permanence, and potential off-target effects.2 These complexities are not insurmountable, but they underscore the need for rigorous translational research, standardization of delivery methods, and careful selection of therapeutic targets.
As the field advances, continued refinement of both ligands and delivery strategies will be necessary to ensure that the promise of DREADDs can be realized in safe, effective, and clinically meaningful ways.
Toward Therapeutic Use: DREADDs as Clinical Tools
While DREADDs were originally developed as research tools, their evolution over the past two decades has positioned them as serious contenders in the expanding toolkit of clinical neuromodulation and gene therapy. The ability to selectively and reversibly modulate specific cell populations using systemically administered ligands aligns well with the goals of precision medicine. Encouraged by encouraging preclinical outcomes, researchers are now working to bridge the gap between laboratory application and therapeutic deployment.
The strongest case for DREADD-based therapy comes from recent studies in Parkinson’s disease models, where selective activation of D1 receptor–expressing medium spiny neurons using a humanized Gs-coupled DREADD (hM3Ds) produced measurable motor improvements.11,12 These findings suggest that chemogenetic stimulation of specific pathways can achieve therapeutic effects without the need for continuous deep brain stimulation or the side effects associated with systemic dopaminergic agents. The localized nature of DREADD activation also reduces the likelihood of widespread off-target effects, providing a compelling alternative or complement to more invasive neuromodulation strategies.
Compared with other neuromodulatory platforms — such as optogenetics, focused ultrasound, or brain–machine interfaces (BMIs) —DREADDs offer distinct advantages in terms of clinical feasibility. Optogenetics, while precise, requires implanted fiberoptics and high-energy light sources, making it difficult to translate beyond small-animal models. Focused ultrasound is noninvasive but limited in spatial resolution and depth. BMIs enable real-time feedback and closed-loop control, but they remain technically complex and often rely on physical implants. DREADDs, by contrast, rely on gene delivery and orally or intravenously administered ligands, making them more amenable to repeat dosing, outpatient use, and integration with existing pharmacological frameworks.
Looking ahead, the range of potential clinical targets for DREADDs is broad. Neurological indications, such as Parkinson’s disease, epilepsy, and treatment-resistant depression are obvious starting points given the existing body of preclinical data and the centrality of circuit dysfunction in these disorders. Peripheral targets are also gaining traction. The development of HCAD receptors for immune modulation illustrates how DREADDs could be used to treat inflammatory or autoimmune conditions without affecting central pathways.14 Other potential applications include metabolic regulation in the liver and pancreas, cardiac rhythm modulation, and gastrointestinal motility control.15 The flexibility of the platform also makes it well-suited to address pediatric or rare diseases, where conventional therapies may be ineffective, unavailable, or poorly tolerated.
Translating DREADDs from bench to bedside will require overcoming several practical and regulatory hurdles. One critical requirement is the production of good manufacturing practice (GMP)-grade viral vectors with validated quality, purity, and lot-to-lot consistency. Parallel efforts must focus on the development and approval of ligands that meet human safety standards, with optimized pharmacokinetics and minimal off-target activity. Longitudinal studies in non-human primates will be essential to demonstrate durable expression, predictable responses, and long-term safety — criteria already being explored in recent research.12
Finally, regulatory approval for a DREADD-based therapy will likely involve a dual-pathway strategy, treating the receptor and ligand as a combined gene-plus-drug product. This hybrid regulatory model may draw from existing gene therapy and combination product frameworks, but it will require proactive engagement with agencies and careful definition of endpoints, safety margins, and reversibility protocols.
The path to clinical adoption is complex, but the tools, precedent, and momentum are aligning. As gene delivery systems and synthetic ligand design continue to mature, DREADDs may soon emerge not only as a staple of neuroscience labs but also as precision medicines for some of the most difficult-to-treat conditions.
The Road Ahead: Future Directions in DREADD Technology
As DREADDs approach the threshold of clinical translation, ongoing innovation continues to expand the boundaries of what chemogenetic modulation can achieve. The next phase of development is focused on refining the core components of the platform — ligands, vectors, and targeting strategies — while integrating DREADDs with complementary technologies to enable smarter, safer, and more personalized therapies.
One of the most pressing needs is the development of next-generation ligands with improved specificity, stability, and pharmacokinetic profiles. Ideal agonists would be non-metabolizable, free of central nervous system activity outside of DREADD activation, and exhibit predictable, tunable dosing behavior. While C21 represents a step forward compared with CNO and clozapine, recent findings regarding its off-target effects underscore the need for further chemical refinement. Ongoing ligand development will likely draw from medicinal chemistry, computational docking, and high-throughput screening to generate candidates suitable for human use.
Vector optimization is another critical area of progress. While AAVs remain the dominant delivery vehicle, efforts are underway to develop new capsid variants with enhanced tropism, reduced immunogenicity, and the ability to cross challenging biological barriers such as the blood-brain barrier. These engineered AAVs may be combined with synthetic promoters or recombinase-dependent cassettes to enable highly cell-specific expression. Beyond traditional vectors, advances in non-viral delivery and transient expression systems could further expand the scenarios in which DREADDs can be deployed.
Integration with real-time feedback systems is also on the horizon. By combining DREADDs with biosensors that detect disease-relevant signals — such as calcium flux, metabolic markers, or electrophysiological activity — it may be possible to construct closed-loop circuits that autonomously modulate activity based on biological need. Such systems would represent a significant leap in therapeutic precision, enabling temporally responsive intervention in dynamic conditions like seizures or arrhythmias.
Looking even further ahead, DREADDs could be fused with programmable gene-editing platforms such as CRISPR/Cas or RNA-editing systems to allow activity-dependent control of gene expression. This would transform DREADDs from purely modulatory tools into intelligent therapeutic nodes capable of sensing, computing, and responding to cellular states. The convergence of chemogenetics with synthetic biology could ultimately yield hybrid systems that operate like logic gates within living tissue.
Realizing these possibilities will require parallel advances in regulatory science and commercialization frameworks. The combination of gene and drug products presents novel challenges for approval pathways, especially when therapies are tailored to rare diseases or individualized tissue targets. Ethical considerations will also grow in importance, particularly as DREADDs move into pediatric, cognitive, or behavioral indications. Public perception, access equity, and post-market surveillance will be essential components of responsible deployment.2,3
DREADDs have already transformed neuroscience research. What comes next is a coordinated effort to transform them into clinically robust, ethically grounded, and technologically integrated tools for the future of medicine.
Conclusion: A Chemogenetic Revolution in the Making
DREADDs have rapidly progressed from niche research tools to one of the most promising modalities for precision control of cellular activity. Their unique combination of spatial, temporal, and molecular specificity — coupled with noninvasive activation — makes them exceptionally well-suited to interrogate and modulate complex biological systems. What began as an experimental technique for dissecting neural circuits is now evolving into a platform with clear therapeutic potential across neurological, psychiatric, immunological, and metabolic diseases.
The continued refinement of DREADD technology — from high-specificity ligands and immune-stealth vectors to synthetic receptor libraries and closed-loop control systems — is expanding the boundaries of what chemogenetics can accomplish. Parallel progress in preclinical modeling, delivery strategies, and regulatory science is paving the way for clinical translation, with several proof-of-concept studies already demonstrating disease-modifying effects in animal models.
As challenges related to safety, specificity, and scalability are addressed, DREADDs are poised to become a central component of future neuromodulation therapies. Their modular design, adaptability to diverse biological contexts, and compatibility with existing gene delivery infrastructures position them uniquely within the landscape of emerging biologics and gene therapies. With thoughtful innovation and careful translation, DREADDs could help usher in a new era of programmable, patient-specific medicine — one in which biology itself becomes the interface for therapeutic control.
References
1. Roth, Bryan L. “DREADDs for Neuroscientists.” Neuron. 89: 683–694 (2017).
2. “The Potential of DREADDs: A Game-Changer in Neuroscientific Research.” biohippo. 28 Mar. 2024.
3. Chen, Yuerong MM, et al. “Research trends in chemogenetics for neuroscience in recent 14 years: A bibliometric study in CiteSpace.” Medicine. 102: e35291 (2023).
4. Vardy, Eyal, et al. “A New DREADD Facilitates the Multiplexed Chemogenetic Interrogation of Behavior.” Neuron. 86: 936–946 (2015).
5. Lee, Hyeong-Min, Patrick M Giguere, and Bryan L Roth. “DREADDs: novel tools for drug discovery and development.” Drug Discov. Today. 19: 469–473 (2013).
6. Smith, Kyle, et al. “DREADDs: Use and Application in Behavioral Neuroscience.” Behavioral Neuroscience. 130: 137–155 (2016).
7. “Scientists Elucidate How DREADD Technology Highjacks Neuronal Activity.” UNC Health and UNC School of Medicine Newsroom. 30 Nov. 2022.
8. Mueller, John-Sebastian, Fabio Cesar Tescarollo, and Hai Sun. “DREADDs in Epilepsy Research: Network-Based Review.” Front. Mol. Neurosci. 6 Apr. 2022.
9. Whissell, Paul D, Sarasa Tohyama, and Loren J. Martin. “The Use of DREADDs to Deconstruct Behavior.” Front. Genet. 16 May 2016.
10. Zhu, Hu and Bryan L Roth. “DREADD: A Chemogenetic GPCR Signaling Platform.” International Journal of Neuropsychopharmacology. 18: pyu007 (2014).
11. Zhang, Qi, et al. “A humanized Gs-coupled DREADD for circuit and behavior modulation.” Front. Cell Neurosci. 8 Apr. 2025.
12. Nagai, Yuji, et al. “Longitudinal assessment of DREADD expression and efficacy in the monkey brain.” eLife. 2 Apr. 2025.
13. Goutaudier, Raphaël, et al. “DREADDs: The Power of the Lock, the Weakness of the Key. Favoring the Pursuit of Specific Conditions Rather than Specific Ligands.” eNeuro. 6:ENEURO.0171-19.2019 (2019).
14. Miyake, Takahito, et al. “Size-reduced DREADD derivatives for AAV-assisted multimodal chemogenetic control of neuronal activity and behavior.” Cell Reports Methods. 4: 100881 (2024).
15. Lee, Sophie H, Aline Mak, and Mark HG Verheijen. “Comparative assessment of the effects of DREADDs and endogenously expressed GPCRs in hippocampal astrocytes on synaptic activity and memory.” Front. Cell Neurosci. 26 Mar. 2023.
16. Dobrzanski, Grzegorz and Malgorzata Kossut. “Application of the DREADD technique in biomedical brain research.” Pharmacological Reports. 69: 213–221 (2017).