Fact-checked by Grok 2 weeks ago

In vivo

In vivo (Latin for "within the living") refers to biological experiments, observations, or processes conducted within a whole living , such as animals or humans, to study physiological responses in a natural context. This approach contrasts with in vitro methods, which occur outside the in artificial settings like cultures or test tubes, often lacking the full complexity of systemic interactions. In vivo studies are fundamental in fields like and , where they evaluate , safety, and mechanisms by capturing , , , and dynamics that isolated systems cannot replicate. These experiments typically follow initial screening to confirm promising candidates before advancing to preclinical animal models, providing data on potential toxicities and therapeutic effects in intact biological systems. Their physiological relevance makes them critical for bridging findings to clinical outcomes, as evidenced by their role in reducing failure rates in trials through early identification of inefficacy or adverse reactions. While in vivo research has driven advancements in vaccine development, cancer therapies, and infectious disease treatments by revealing organism-level responses, it faces challenges including ethical concerns over animal use and translational limitations from species differences. Nonetheless, empirical evidence underscores its necessity, as alternatives like in silico modeling or organoids often fail to fully predict whole-body pharmacokinetics or immune interactions observed only in living subjects. Ongoing refinements, such as advanced imaging for non-invasive monitoring, continue to enhance its precision and reduce animal requirements without compromising causal insights into disease mechanisms.

Definition and Core Concepts

Etymology and Precise Meaning

The term in vivo derives from Latin, where in means "in" or "within" and is the ablative form of vivus, meaning "alive" or "living," yielding a of "within the living" or "in a living [thing]." This phrase entered scientific lexicon in the late amid advances in , initially to distinguish biological phenomena observed in intact organisms from those replicated in isolated systems. In biomedical research, in vivo precisely refers to experiments, tests, or processes conducted within a whole, living —such as animals, humans, or other intact biological systems—to capture holistic physiological responses, including intercellular interactions, metabolic pathways, and environmental influences absent in controlled isolates. This contrasts with artificial or partial simulations by emphasizing the organism's native milieu, where variables like blood flow, immune modulation, and can alter outcomes in ways not replicable externally. For instance, efficacy assessments in vivo account for , , and potential toxicities across systems, providing data on real-world applicability that informs clinical translation.

Distinctions from In Vitro, In Silico, and Ex Vivo

In vivo experimentation entails biological processes and interventions performed directly within a living , such as mammals, , or microorganisms, thereby capturing dynamic interactions across physiological systems including circulation, immune responses, and metabolic pathways. This approach preserves the organism's holistic complexity, enabling observation of emergent phenomena like drug distribution to target sites and off-target effects that isolated systems cannot replicate. In vitro methods, by comparison, isolate cellular or molecular components—such as cultured cells or enzymes—in artificial environments like multi-well plates or bioreactors, facilitating and mechanistic dissection under controlled conditions devoid of organismal influences. While permitting precise manipulation of variables, such as substrate concentrations or genetic knockouts, in vitro assays frequently yield results that diverge from in vivo outcomes due to the absence of architecture, vascularization, and endocrine signaling, contributing to translational failures estimated at over 90% in early pipelines. Ex vivo techniques involve extracting viable tissues or organs from the donor organism and subjecting them to experimental conditions outside the body, often with perfusion or culture media to sustain short-term functionality, as in isolated heart preparations or skin biopsies. This intermediary paradigm retains native multicellular organization and extracellular matrices absent in standard in vitro setups, yet imposes limitations on duration—typically hours to days—and scalability, precluding long-term adaptive responses or systemic feedback loops inherent to in vivo contexts. In silico modeling employs algorithms, databases, and simulations on computational platforms to forecast biological behaviors, such as via or pharmacokinetic profiles through quantitative structure-activity relationship (QSAR) analyses, without any biological material. These virtual approaches excel in hypothesis generation and optimization, processing vast datasets rapidly—for instance, screening millions of compounds prior to synthesis—but their predictions hinge on parameterized models that may inadequately account for biological variability or nonlinear interactions, as demonstrated by discrepancies between simulated and empirical endpoints in regulatory validations. Collectively, these distinctions underscore in vivo's superior fidelity for in intact systems, albeit at higher ethical, logistical, and financial costs compared to the reductionist alternatives.

Historical Development

Early Physiological Foundations (19th-early 20th Century)

The foundations of in vivo physiological research in the were laid through systematic experimentation on living animals, primarily via , which allowed direct observation of dynamic bodily processes unattainable through postmortem or methods. François Magendie (1783–1855), a French physiologist, pioneered this approach by conducting unanesthetized dissections on animals such as dogs and puppies to elucidate neural functions. In 1822, he demonstrated the distinct roles of spinal nerve roots, confirming that dorsal roots transmit sensory impulses while ventral roots convey motor signals, resolving a prior dispute with and establishing empirical criteria for physiological inference. His 1809 experiments injecting plant extracts into living animals also marked the inception of experimental , revealing emetic and paralytic effects that informed therapeutic applications. Claude Bernard (1813–1878), Magendie's student, advanced in vivo methods into a rigorous scientific framework, advocating hypothesis-testing through controlled live-animal studies over speculative . In his 1865 Introduction to the Study of Experimental , Bernard emphasized the necessity of in vivo observation to capture organismal responses, critiquing reductionist alternatives and introducing the concept of milieu intérieur—the regulated internal environment essential for life, later foundational to . Key discoveries included the pancreas's role in fat digestion (via canine experiments in the 1840s) and the liver's synthesis from non-carbohydrate sources (1850s curare and nutrient infusion studies on rabbits and dogs), which revealed metabolic regulation in intact systems. Bernard's insistence on quantitative, repeatable in vivo protocols elevated from descriptive to causal science, influencing global laboratories. Entering the early 20th century, in vivo techniques expanded to integrate behavioral and neural dynamics, as seen in Ivan Pavlov's (1849–1936) chronic fistula preparations on dogs, which enabled long-term monitoring of salivary and gastric secretions under natural conditions. Awarded the 1904 Nobel Prize, Pavlov's work quantified neural-glandular reflexes, demonstrating cephalic phase digestion through vagal stimulation and laying groundwork for conditioned responses via repeated in vivo pairings of stimuli with feeding. Concurrently, Charles Sherrington (1857–1952) refined in vivo neurophysiology through spinal and decerebrate preparations in cats and monkeys, identifying reciprocal inhibition in reflexes (late 1890s) and coining "synapse" in 1897 to describe junctional transmission observed in living preparations. These advancements underscored in vivo's superiority for revealing integrative mechanisms, bridging cellular events to organismal function amid emerging antisepsis that reduced procedural artifacts.

Post-WWII Expansion in Biomedical Research

Following , the United States government substantially increased funding for biomedical , transforming institutions like the (NIH) from modest operations into a primary grant-making agency supporting extramural studies. Pre-war NIH appropriations stood at approximately $700,000 annually, rising to $3.4 million by war's end and expanding further in the postwar period to fuel a broader enterprise. This expansion built directly on wartime initiatives, such as the Committee on Medical Research (CMR), which coordinated cross-sectoral efforts to advance medical technologies amid military needs, laying groundwork for sustained peacetime innovation in and disease treatment. The postwar emphasis privileged empirical validation through , accelerating in vivo approaches to capture physiological complexities unattainable in isolated cellular or computational models. A key driver of this growth was the proliferation of animal models in biomedical investigations, with laboratory usage surging to meet demands for , infectious disease modeling, and pharmacological trials. Post-1945, animal experimentation volumes rose markedly due to expanded university programs and pipelines, with mice and rats becoming staples for studying radiation effects, cancer induction, and vaccine responses—exemplified by large-scale exposures to that amplified breeding and utilization scales. By the , this infrastructure enabled breakthroughs like the in vivo validation of penicillin derivatives and early antimalarials, where whole-organism testing confirmed , immune interactions, and adverse effects in contexts mirroring . Human in vivo research also burgeoned through clinical trials and observational cohorts, informed by ethical reforms stemming from the 1947 , which mandated voluntary consent following wartime atrocities. The NIH's role in funding such studies facilitated pivotal trials, including the 1954 Salk field test involving over 1.8 million children, which relied on prior model validations to assess and in living subjects. This era's causal focus—prioritizing interventions' systemic impacts over reductionist alternatives—underscored in vivo methods' indispensability, despite emerging debates over and human subject protections that prompted guidelines like the 1964 . Overall, these developments entrenched in vivo paradigms as foundational to evidence-based , yielding causal insights into disease mechanisms and therapeutic outcomes unattainable otherwise.

Methodological Techniques

Animal Model Implementation

Rodents, particularly mice (Mus musculus) and rats (Rattus norvegicus), serve as primary animal models in in vivo research due to their genetic homology to humans (approximately 99% shared in mice), short reproductive cycles, and ease of genetic manipulation. Strains such as , , and Wistar are commonly selected for inbred consistency in physiological responses, while outbred strains like Swiss Webster provide variability mimicking human populations. Implementation begins with procurement or breeding of specific pathogen-free animals, followed by housing in controlled environments adhering to standards like those from the Association for Assessment and Accreditation of Laboratory Animal Care (AAALAC). Disease modeling in these animals employs induced, genetic, or spontaneous techniques to replicate human pathologies. Chemical induction, such as (STZ) administered intraperitoneally at 65 mg/kg body weight in Wistar rats to induce , confirms (≥10 mmol/L) within 72 hours post-injection. Genetic models utilize /Cas9-mediated editing to create knockouts or transgenics, as in mouse models with mutations in , Tp53, and Lkb1 genes for simulation, enabling study of gene-specific effects in a whole-organism context. Surgical orthotopic implantation, such as injecting (10² CFU/10 μL) into rat tibial metaphysis for , or creating burn wounds with a 100°C aluminum rod applied for 15 seconds, further mimics localized diseases. Interventions involve precise administration routes tailored to , including oral gavage, intravenous, subcutaneous, or intraperitoneal injections, often under like (4% induction, 2% maintenance) or ketamine/ combinations to minimize stress. For efficacy testing, such as in antimalarial drug studies, Plasmodium-infected mice receive prophylactic, suppressive (e.g., Peter's 4-day test), or curative dosing regimens, with humanized models like NOD-scid IL2Rγ null strains accommodating human-specific pathogens. Monitoring encompasses non-invasive imaging (e.g., MRI for defects) and behavioral assays, alongside analgesia like (0.05 mg/kg) to comply with welfare protocols. Outcomes are assessed through multimodal endpoints, including blood and tissue sampling analyzed via high-performance liquid chromatography-electrospray ionization-mass spectrometry (HPLC-ESI-MS/MS) for drug levels, for toxicity, and parasitemia quantification via or for efficacy. Biomechanical testing evaluates repair, while survival metrics and morbidity scores predict translational relevance, as in rasH2 models for carcinogenicity that reduce required sizes compared to traditional two-year studies. Necropsy follows humane endpoints, with data integrated to inform preclinical progression.

Human Clinical and Observational Studies

Human clinical trials represent a critical phase of in vivo , involving the administration of interventions to living subjects to assess , , and physiological responses within the body's integrated systems. These trials typically progress through four phases: Phase I focuses on and dosage in small groups (20-100 healthy volunteers or patients), establishing and tolerability; Phase II evaluates and side effects in larger cohorts (hundreds); Phase III compares interventions against standards in thousands of participants via randomized controlled designs; and Phase IV monitors long-term effects post-approval. Randomization and blinding minimize bias, while ethical oversight by institutional review boards ensures informed consent and risk-benefit balance, as mandated by the 1964 and U.S. Title 21. Observational studies in humans, distinct from interventional trials, capture real-world in vivo data without manipulating variables, relying on cohort (prospective tracking of exposed vs. unexposed groups), case-control (retrospective comparison of outcomes), or cross-sectional designs. For instance, the Framingham Heart Study, initiated in 1948, has longitudinally observed cardiovascular risk factors in over 15,000 participants across generations, revealing causal links like hypertension to heart disease through multivariate analysis adjusted for confounders. These studies excel in identifying associations for rare events or long latency outcomes, such as the Nurses' Health Study (1976 onward, n>280,000 women) linking diet and lifestyle to chronic disease incidence via hazard ratios and population attributable risks. However, they are prone to confounding and reverse causation, necessitating techniques like propensity score matching or instrumental variable analysis for causal inference. Integration of clinical and observational data enhances in vivo validation, as seen in where post-marketing surveillance (e.g., FDA's Sentinel System, analyzing electronic health records from millions) detects rare adverse events missed in trials, such as the 1-2 per million incidence of Guillain-Barré syndrome post-vaccination identified via Bayesian methods. Hybrid approaches, like pragmatic trials embedding in routine care, bridge gaps, exemplified by the SPRINT trial (2015, n=9,361 hypertensives) demonstrating intensive control (<120 mmHg systolic) reduced cardiovascular events by 25% (HR 0.75, 95% CI 0.64-0.89) compared to standard targets. Despite strengths in capturing holistic human physiology—including immune, metabolic, and behavioral interactions—limitations persist, including generalizability biases from underrepresentation of minorities (e.g., only 5-10% non-white participants in many U.S. trials pre-2020) and high attrition rates (up to 20-30% in long-term studies). Regulatory bodies like the EMA emphasize diverse recruitment via adaptive designs to mitigate these. Advanced in vivo human methodologies incorporate biomarkers and imaging for mechanistic insights, such as positron emission tomography (PET) in oncology trials tracking tumor metabolism via FDG uptake, correlating with progression-free survival in Phase II studies of targeted therapies. Real-time monitoring via wearables and digital endpoints, piloted in COVID-19 vaccine trials (e.g., Pfizer-BioNTech, 2020, n=44,000), quantified symptom resolution and antibody titers, yielding 95% efficacy against symptomatic infection (95% CI 90.3-97.6). These approaches underscore in vivo human studies' role in causal realism, prioritizing empirical outcomes over preclinical proxies, though source credibility varies—industry-sponsored trials report 20-30% higher efficacy estimates than independent ones, per meta-analyses, warranting scrutiny of funding disclosures.

Non-Mammalian Organism Applications

Non-mammalian organisms, including invertebrates like Drosophila melanogaster and Caenorhabditis elegans and vertebrates such as zebrafish (Danio rerio), enable in vivo experimentation through their genetic amenability, rapid reproduction, and physiological similarities to higher organisms in key pathways. These models facilitate whole-organism studies of development, genetics, and pharmacology, often bypassing mammalian ethical hurdles while allowing real-time imaging and genetic manipulation. In Drosophila melanogaster, in vivo techniques involve large-scale genetic screens to dissect gene functions, as demonstrated by a 2024 study testing over 5,000 human genes for roles in signaling pathways via CRISPR-induced mutations and phenotypic assays. Flies' short 10-day generation time supports forward and reverse genetics, modeling human diseases like neurodegeneration and cardiovascular disorders through tissue-specific expression of mutant transgenes. Behavioral assays, such as locomotion tracking, quantify in vivo effects of interventions on neural circuits. Caenorhabditis elegans supports in vivo neuroscience and aging research via its 302-neuron connectome, enabling precise optogenetic and chemogenetic manipulation. Studies reveal age-related neuronal changes, including soma volume reduction and neurite outgrowth increases by up to 20% in aged worms, tracked via quantitative imaging from day 1 to day 21 of adulthood. High-throughput screens assess compound effects on lifespan and motility, identifying anti-aging interventions that extend mean lifespan by 15-30% in wild-type strains. Zebrafish in vivo applications leverage embryonic transparency for non-invasive imaging of vascular and neural dynamics, with larvae used in drug discovery screens processing thousands of compounds weekly. In toxicology, embryos exposed to nanoparticles from 5 days post-fertilization show dose-dependent survival rates, predicting mammalian toxicity with 70-80% concordance in cardiovascular assays. Disease models, such as induced tuberculosis via Mycobacterium marinum injection, validate anti-infective efficacy, reducing bacterial burden by 50% in responsive larvae. Genetic tools like TALENs and CRISPR enable targeted knockouts, modeling neurodevelopmental disorders with behavioral phenotyping at 96 hours post-fertilization.

Key Applications

Drug Development and Toxicology Testing

In vivo testing constitutes a cornerstone of preclinical drug development, enabling evaluation of a compound's absorption, distribution, metabolism, excretion (ADME), pharmacodynamics, and potential toxicity in intact biological systems. These studies typically involve administering candidate drugs to animal models, such as rodents (e.g., rats and mice) and non-rodents (e.g., dogs or non-human primates), to identify dose-response relationships, therapeutic indices, and off-target effects that in vitro methods cannot replicate due to the absence of systemic interactions. Toxicology testing in vivo encompasses multiple study types designed to detect adverse effects across exposure durations and endpoints. Acute toxicity studies expose animals to high doses over short periods (e.g., 14 days) to determine median lethal dose (LD50) and immediate hazards, while subchronic and chronic studies (lasting 90 days to two years) assess cumulative organ damage, carcinogenicity, and genotoxicity in models like the two-year rodent bioassay. Reproductive and developmental toxicity (DART) studies evaluate impacts on fertility, embryofetal development, and offspring in multi-generational rodent cohorts. Safety pharmacology core batteries test effects on cardiovascular, respiratory, and central nervous systems, often via telemetry in conscious animals. Regulatory frameworks have historically mandated in vivo data for advancing to human trials; prior to amendments, the U.S. FDA required toxicity studies in two mammalian species—one rodent and one non-rodent—for investigational new drug (IND) applications to establish safe starting doses via no-observed-adverse-effect levels (NOAELs). Although the FDA Modernization Act 2.0, enacted December 29, 2022, eliminated the compulsory use of animals by permitting alternatives like organoids and computational models when supported by evidence, in vivo studies persist as the gold standard for validating complex toxicities, comprising approximately 10% of all animal experiments in drug discovery. In practice, in vivo toxicology informs go/no-go decisions, with findings guiding formulation adjustments or termination of unviable candidates; for instance, repeat-dose studies in GLP-compliant facilities monitor clinical pathology, histopathology, and biomarkers to predict human risks, as evidenced by their role in de-risking over 90% of development-stage compounds before clinical entry. These applications underscore in vivo's utility in bridging preclinical gaps, despite ongoing efforts to integrate non-animal approaches for efficiency and ethics.

Genetic and Developmental Studies

In vivo genetic studies employ living model organisms to elucidate gene functions, regulatory networks, and epistatic interactions within intact physiological environments, revealing systemic effects unattainable through isolated cellular assays. Common models include the mouse (Mus musculus), fruit fly (Drosophila melanogaster), zebrafish (Danio rerio), and nematode (Caenorhabditis elegans), selected for their genetic amenability, rapid reproduction, and evolutionary conservation of developmental pathways with vertebrates. These organisms facilitate forward and reverse genetic screens, such as in vivo RNAi to systematically silence genes and observe phenotypic outcomes during development. Gene knockout techniques, pioneered through homologous recombination in mouse embryonic stem cells, have been instrumental in assigning developmental roles to specific genes, with targeted disruptions often yielding embryonic lethality in about 15% of cases, indicating indispensable functions in embryogenesis or organogenesis. For instance, knockouts of tumor suppressor genes like Brca1 and Brca2 demonstrate early embryonic arrest linked to proliferation defects and p53 pathway activation, underscoring causal roles in cellular viability and genomic stability during gestation. Conditional knockouts, using tissue-specific Cre-loxP systems, further refine these analyses by bypassing lethality to probe gene actions in postnatal or stage-specific contexts, as seen in excitatory versus inhibitory neuron disruptions revealing behavioral phenotypes. Advancements in CRISPR-Cas9 have expanded in vivo manipulation capabilities, enabling precise multiplexed editing directly in embryos or adult tissues to model developmental disorders and validate causal variants. This technology supports homology-independent targeted integration for correcting mutations in patient-derived induced pluripotent stem cell models transplanted into organisms, bridging genetic findings to organismal phenotypes. In developmental biology, in vivo approaches capture dynamic processes like neuronal migration and apoptosis, as evidenced by estrogen receptor β knockouts impairing late embryonic brain development. Quantitative imaging of live embryos complements these genetic perturbations, providing spatiotemporal resolution of morphogenetic events and tissue interactions essential for understanding congenital anomalies. Such studies highlight in vivo's superiority for discerning holistic gene-environment interplay, where in vitro models often fail to recapitulate emergent properties like epigenetic remodeling or intercellular signaling cascades driving organ formation. Population-based genetic backgrounds in diverse mouse strains further mitigate artifacts from inbred lines, accelerating translation to human developmental genetics. Despite interspecies extrapolation challenges, these methods have conserved disease genes across models, informing diagnostics and therapies for rare disorders.

Disease Modeling and Therapeutic Validation

In vivo disease modeling utilizes living organisms, primarily rodents and other mammals, to replicate human disease states, capturing complex physiological interactions absent in isolated cellular systems. These models facilitate the examination of disease onset, progression, and multifaceted responses to perturbations, grounded in the organism's integrated biology. Rodent models predominate due to their short generation times, genetic tractability, and partial homology to human physiology, enabling targeted manipulations like gene knockouts or transgenics to induce pathology. For (AD), transgenic mice harboring mutations in the amyloid precursor protein (APP) and presenilin 1 (PS1) genes exhibit amyloid-beta plaque accumulation, tau hyperphosphorylation, and synaptic loss, mirroring key AD neuropathological features observed in postmortem human brains. Similarly, in type 1 diabetes modeling, administration of streptozotocin to rodents selectively destroys pancreatic beta cells, inducing hyperglycemia and insulin dependence akin to autoimmune beta-cell loss in humans. Cancer disease modeling in vivo often employs xenograft or orthotopic implantation of human tumor cells into immunocompromised mice, allowing real-time monitoring of tumor growth, vascularization, and metastasis under physiological conditions. These setups reveal systemic effects like immune evasion or organ-specific dissemination, which in vitro assays overlook. For therapeutic validation, in vivo platforms test drug candidates for efficacy, biodistribution, and off-target toxicities; for example, patient-derived tumor xenografts have identified novel agents effective against high-risk neuroblastomas by screening compounds in vivo post-in vitro hits, correlating tumor regression with molecular target engagement. In AD therapeutic pipelines, rodent models have validated anti-amyloid interventions, such as monoclonal antibodies reducing plaque burden, though human translation varies due to species-specific protein processing differences. Despite these applications, in vivo validation's predictive power for human outcomes is limited, with only about 10-12% of preclinical candidates advancing successfully from animal testing to Phase I clinical trials, attributable to interspecies physiological variances and incomplete disease recapitulation. Peer-reviewed analyses emphasize fit-for-purpose validation criteria—face validity (phenotypic similarity), construct validity (etiological accuracy), and predictive validity (therapeutic translatability)—to enhance reliability, yet systemic failures persist, underscoring the need for complementary human-relevant assays. In diabetes drug development, rodent models successfully predicted insulin analogs' glycemic control but faltered in forecasting human immune responses to novel peptides. Overall, while in vivo modeling provides causal insights into therapeutic mechanisms via longitudinal tracking of biomarkers and endpoints, its role is preparatory, informing dose escalation and safety profiles before human testing.

Empirical Advantages

Holistic Physiological Interactions

In vivo studies facilitate the analysis of interconnected physiological processes across an intact organism, including inter-organ signaling via circulatory systems, endocrine feedback, and immune modulation, which collectively maintain homeostasis and respond to perturbations in ways unattainable through isolated cellular or tissue-based in vitro approaches. These models capture dynamic, bidirectional communications—such as cytokine-mediated cross-talk between liver and adipose tissue during metabolic stress or neural-immune interactions in inflammation—revealing emergent properties like adaptive resistance or compensatory mechanisms that arise from whole-body integration. Empirical evidence from rodent models of chronic diseases demonstrates that such systemic interplay influences outcomes like insulin sensitivity, where hepatic glucose production modulates pancreatic beta-cell function in real time, underscoring the causal realism of organism-level experimentation over reductionist alternatives. A key advantage lies in replicating the spatiotemporal complexity of physiological responses, where in vivo imaging techniques, such as fluorescence microscopy in live animals, track molecular events alongside tissue-level changes and behavioral correlates. For example, in cancer research, in vivo observations reveal how tumor cells interact with stromal and vascular components to drive metastasis, including extracellular matrix remodeling and angiogenic signaling, processes distorted in vitro due to absent vascular perfusion and immune surveillance. This holistic framework enhances predictive validity for therapeutic interventions, as evidenced by longitudinal studies showing biodistribution patterns and pharmacokinetic interactions that inform dosing regimens, reducing translational failures from preclinical to clinical stages. Furthermore, in vivo paradigms enable the study of feedback loops and redundancy inherent to living systems, such as the regulating stress responses across endocrine, neural, and peripheral tissues. Disruptions in these networks, observable only in whole-organism contexts, highlight limitations of in vitro models, which often overlook dilution effects from blood volume or multi-compartmental pharmacokinetics, leading to overestimation of efficacy or toxicity. Quantitative data from animal cohorts, including survival metrics and biomarker panels, validate these interactions' role in disease modeling, as seen in cardiovascular studies where cardiac output influences renal filtration and vice versa, providing a more accurate basis for causal inference than compartmentalized assays.

Superior Predictive Accuracy for Systemic Effects

In vivo methodologies demonstrate superior predictive accuracy for systemic effects compared to in vitro or computational alternatives, as they replicate the integrated physiological dynamics absent in isolated cellular or tissue models. Systemic effects, encompassing pharmacokinetics such as absorption, distribution, metabolism, and excretion (ADME), as well as multi-organ crosstalk and compensatory responses, emerge from whole-organism interactions that in vitro systems inherently fail to capture due to their static, non-vascularized environments. For example, hepatic metabolism of prodrugs into active or toxic metabolites, followed by redistribution to distant sites like the heart or kidneys, can only be reliably assessed in living models where blood flow, enzyme induction, and immune modulation occur dynamically. Empirical data underscore this advantage: analyses of preclinical toxicity testing reveal that non-animal models, including in vitro assays and in silico predictions, cannot forecast systemic effect levels—such as no-observed-adverse-effect levels (NOAELs)—with accuracy exceeding that of animal studies. Concordance rates for systemic toxicity between multi-species in vivo data and human outcomes reach 71% overall, with non-rodent models achieving 63%, outperforming in vitro extrapolations that often overestimate potency or miss secondary toxicities due to lacking systemic context. This predictive edge is evident in regulatory frameworks, where in vivo testing remains indispensable for identifying delayed or cumulative systemic risks, such as chronic inflammation or endocrine disruption, which isolated cell cultures predict poorly. Furthermore, in vivo approaches excel in revealing emergent toxicities from physiological feedbacks, like hypotension-induced renal failure or cytokine storms affecting multiple organs, which in vitro models simulate inadequately without integrated organoid networks or real-time homeostasis. While interspecies physiological variances limit absolute human predictivity, in vivo's holistic framework provides a foundational filter for systemic hazards, reducing false negatives in early drug screening that plague reductionist alternatives. Ongoing refinements, such as physiologically based pharmacokinetic (PBPK) modeling informed by in vivo data, further enhance this accuracy by bridging species gaps for systemic dose-response predictions.

Criticisms and Limitations

Ethical Debates on Animal Welfare

Ethical debates surrounding animal welfare in in vivo research primarily contrast the potential human benefits of biomedical advancements with the documented capacity of animals to experience pain, distress, and psychological harm. Critics, drawing on evidence of sentience in species like rodents and primates, argue that such experiments inflict unnecessary suffering, violating principles of moral consideration for beings capable of negative affective states. For instance, procedures often involve invasive surgeries, toxin exposure, or genetic manipulations that cause acute or chronic pain, with empirical observations confirming behavioral and physiological indicators of stress in laboratory animals. Philosophical opposition frequently invokes rights-based frameworks, positing that animals possess intrinsic value independent of human utility, rendering their exploitation akin to unjust discrimination or "speciesism." Utilitarian critics, such as those emphasizing equal consideration of interests, contend that the aggregate harm—evidenced by statistics like the 49% of 9.3 million regulated procedures in the UK during 2022 causing moderate to severe suffering—outweighs uncertain translational benefits, especially amid advancing alternatives. Abolitionist positions, advanced by organizations questioning the ethical permissibility of non-consensual harm, demand complete cessation, viewing welfare regulations as insufficient palliatives. Defenders maintain that human moral priority, rooted in superior cognitive capacities for autonomy, rationality, and long-term planning, justifies animal use under strict cost-benefit analyses where societal gains—such as vaccines preventing millions of deaths—predominate. Regulatory paradigms like the 3Rs (replacement, reduction, refinement) are cited as evidence of ethical progress, having halved UK procedure numbers over decades to approximately 2.7 million by 2003, with ongoing refinements minimizing distress through analgesia and enriched environments. Proponents argue that without in vivo models, critical systemic insights into disease and therapy would be unattainable, as partial alternatives fail to replicate whole-organism dynamics, and public surveys historically affirm support for regulated research when welfare is prioritized. These debates persist amid declining but persistent animal use, underscoring tensions between anthropocentric imperatives and expanding recognition of animal sentience.

Interspecies Translation Gaps and Failure Rates

Interspecies translation gaps in in vivo research stem from profound biological divergences between model organisms and humans, encompassing genetic, physiological, metabolic, and immunological differences that undermine the predictive validity of preclinical outcomes. These disparities often manifest in divergent drug responses, where efficacy or toxicity observed in animals does not replicate in humans due to variations in receptor binding, enzyme activity, and systemic homeostasis. For instance, species-specific cytochrome P450 enzyme profiles lead to differential drug metabolism, with non-human models frequently metabolizing compounds more rapidly or via absent human pathways, resulting in false positives or negatives for pharmacokinetics. Consequently, approximately 90% of drug candidates that advance past animal testing fail in clinical phases I-III, primarily from lack of efficacy (around 50-60% of failures) or unforeseen safety issues not anticipated in preclinical models. In non-mammalian in vivo models, such as Drosophila melanogaster (fruit fly) and Danio rerio (zebrafish), these gaps are exacerbated by greater phylogenetic distance, yielding even lower translational fidelity compared to mammalian counterparts. Non-mammals typically lack complex mammalian structures like a fully adaptive immune system, specialized organs (e.g., no true bone marrow in flies), or homologous metabolic cascades, limiting their utility for predicting human-specific responses in areas like neuropharmacology or oncology. Empirical data show that while these models facilitate initial high-throughput screening, fewer than 5% of findings from animal studies overall—potentially lower for non-mammals—progress to approved human treatments, with non-mammalian validations rarely yielding direct clinical successes due to incomplete recapitulation of human disease states. For example, zebrafish exhibit accelerated developmental timelines and distinct cardiovascular dynamics, which can mask human-relevant toxicities, as seen in variable responses to cardiotoxicants absent in human orthologs. Notable case studies illustrate these failures: thalidomide, initially overlooked for teratogenicity in rodent models due to metabolic differences, caused severe human birth defects; similarly, species variances in hepatotoxicity, such as differing sensitivities to compounds like carbofuran across mammals and potentially wider in non-mammals, highlight how in vivo results can mislead toxicology assessments. In cancer drug development, over 93% of candidates succeeding in animal models, including non-mammalian screens, fail early human trials, underscoring systemic inaccuracies from interspecies biodistribution and effector mechanisms. Despite refinements like multi-species testing, concordance rates for toxicity remain modest (e.g., positive predictive values below 50% for certain endpoints between rodents and humans, likely poorer for non-mammals), prompting calls for adjunct human-based assays to bridge these gaps.

Resource and Scalability Constraints

In vivo experiments demand substantial financial resources, with costs often exceeding those of in vitro alternatives due to the need for specialized animal housing, veterinary care, and regulatory compliance. For instance, a rat developmental toxicity test can cost approximately $50,000, compared to $15,000 for an in vitro rat limb bud test. Base rates for rodent pharmacokinetics studies range from $257 for mice to $1,100 for rats, excluding per-animal housing and procedural fees that can add $40–$50 per animal and escalate total study expenses into tens of thousands of dollars. These expenditures are compounded by infrastructure requirements, such as vivariums and biosafety facilities, which academic and industry labs must maintain amid rising operational costs estimated at billions annually in the U.S. biomedical sector. Scalability is limited by the labor-intensive nature of in vivo work, requiring highly trained personnel for animal handling, dosing, monitoring, and ethical oversight, often governed by good laboratory practice (GLP) standards that mandate dedicated staffing and documentation. A single study may involve teams of technicians, veterinarians, and principal investigators, with hourly tech rates applying to tasks like colony management and experimental execution, further inflating budgets for larger cohorts. Unlike high-throughput in vitro screening, which can process thousands of compounds rapidly, in vivo models struggle to scale due to biological variability, the need for individual animal acclimation, and procedural timelines that preclude parallelization—such as surgical interventions or longitudinal observations spanning weeks to months. These constraints hinder rapid iteration in drug discovery, where resource allocation favors fewer, more controlled in vivo validations over broad exploratory testing, contributing to delays in translating findings to clinical stages. Efforts to optimize, such as streamlined colony production frameworks, aim to reduce animal numbers and costs but cannot fully mitigate the inherent bottlenecks of live-organism experimentation.

Regulatory and Ethical Standards

The 3Rs Principle and Implementation

The 3Rs principle, comprising replacement, reduction, and refinement, serves as a foundational ethical framework for minimizing animal use in scientific research, including in vivo studies conducted within living organisms. Originating from the 1959 publication The Principles of Humane Experimental Technique by William M. S. Russell and Rex L. Burch, the principle emphasizes humane treatment while preserving scientific validity, acknowledging that in vivo models remain essential for investigating complex physiological processes unattainable through non-animal alternatives. Replacement involves substituting animal models with non-animal methods, such as in vitro cell cultures, organoids, or computational simulations, wherever they yield reliable data; for instance, high-throughput screening of compounds using human-derived cells can obviate initial rodent testing in drug discovery pipelines. Reduction entails optimizing experimental designs to extract maximum information from fewer animals, often through statistical power analysis, multiplexing assays, or sharing data across studies, as demonstrated in toxicology where fewer rodents suffice for dose-response curves when historical controls are leveraged. Refinement focuses on alleviating suffering via techniques like analgesia, enriched environments, or early humane endpoints, reducing distress in in vivo procedures such as surgical models of disease. Implementation of the 3Rs in in vivo research is mandated by regulatory frameworks globally, requiring institutional oversight to evaluate and apply these principles prospectively. In the United States, Institutional Animal Care and Use Committees (IACUCs) review protocols under the Animal Welfare Act, enforcing 3Rs compliance by mandating justification for animal use and alternatives assessment, with data from 2022 indicating over 1 million regulated animals annually subjected to such scrutiny. The European Union's Directive 2010/63/EU codifies the 3Rs into law, obliging member states to promote alternatives and report progress, resulting in a reported 10-20% decline in animal numbers for certain toxicity tests between 2013 and 2020 through refined endpoints and partial replacements. In practice, bodies like the UK's National Centre for the Replacement, Refinement and Reduction of Animals in Research (NC3Rs) provide guidelines and tools, such as the Experimental Design Assistant software, which has aided researchers in reducing animal cohorts by up to 30% in neuroscience in vivo models by improving randomization and blinding. Challenges in implementation arise from the irreplaceable role of in vivo studies for systemic effects, where alternatives often fail to replicate whole-organism dynamics, necessitating balanced application: for example, while organ-on-chip systems replace some acute toxicity assessments, chronic in vivo rodent models persist for carcinogenicity testing due to superior predictive power for human outcomes. Ongoing efforts include training programs and funding incentives, with the U.S. National Institutes of Health allocating resources since 2018 to 3Rs innovation, fostering refinements like telemetry for non-invasive monitoring in cardiovascular studies, thereby minimizing surgical interventions. Empirical audits, such as those by the NC3Rs, reveal that rigorous 3Rs adherence correlates with higher data reproducibility, underscoring causal links between ethical minimization and scientific robustness in in vivo experimentation. In the United States, Institutional Animal Care and Use Committees (IACUCs) serve as the primary oversight bodies for in vivo research involving animals, tasked with reviewing protocols, inspecting facilities, and ensuring adherence to welfare standards under the Public Health Service Policy on Humane Care and Use of Laboratory Animals. These committees, mandated for institutions receiving federal funding, include scientists, non-scientists, and community representatives to evaluate the necessity, design, and minimization of animal use. The U.S. Department of Agriculture's Animal and Plant Health Inspection Service (APHIS) enforces compliance through inspections and licensing under the (as amended), which regulates housing, veterinary care, and handling for covered warm-blooded species, excluding purpose-bred rats, mice, and birds. The Health Research Extension Act of 1985 extends oversight to research funded by the Department of Health and Human Services, requiring implementation of the U.S. Government Principles for the Utilization and Care of Vertebrate Animals, which emphasize alternatives to animal use where feasible. Recent developments include the , which eliminated mandatory animal testing for certain drug approvals, permitting non-animal methods like organ chips or computational models while maintaining voluntary adherence to in vivo standards for safety validation. In the European Union, Directive 2010/63/EU establishes a harmonized legal framework for the protection of animals used in scientific procedures, requiring member states to designate competent authorities for authorization, inspections, and enforcement, with mandatory ethical evaluations by multidisciplinary committees prior to project approval. This directive mandates severity classifications for procedures, retrospective assessments for projects exceeding certain thresholds (e.g., involving non-human primates or causing severe pain), and national bans on testing for cosmetics since 2013, while promoting the 3Rs through requirements for alternative method searches and data sharing via databases like the European EURL ECVAM. Transposed into national laws, it applies to all vertebrates and cephalopods, with exemptions only for educational or basic research under strict justification. Internationally, frameworks vary; for instance, the Canadian Council on Animal Care provides voluntary accreditation similar to AAALAC International, which verifies compliance with global standards across over 1,000 institutions in 50 countries as of 2023. Bodies like the International Council for Laboratory Animal Science (ICLAS) promote harmonized ethical guidelines, though enforcement remains jurisdiction-specific, with ongoing debates over aligning U.S. and EU approaches to reduce regulatory burdens while upholding welfare.

Recent Innovations (2020-Present)

In Vivo Gene Editing with CRISPR

In vivo gene editing with enables targeted modifications to DNA directly within living organisms, bypassing the need for ex vivo cell manipulation and reinfusion. This method relies on delivery vehicles such as lipid nanoparticles (LNPs) for systemic hepatic targeting or adeno-associated viral (AAV) vectors for localized applications like the retina, achieving editing efficiencies that can yield therapeutic protein reductions or restorations. Advancements since 2020 have emphasized precision nucleases, reduced off-target cleavage via high-fidelity Cas variants, and scalable manufacturing to address immune evasion and biodistribution challenges inherent to in-body deployment. The inaugural human in vivo CRISPR trial, the phase 1/2 BRILLIANCE study by Editas Medicine, targeted Leber congenital amaurosis type 10 (LCA10) caused by CEP290 mutations. Launched in March 2020, it administered escalating doses of EDIT-101—an AAV5 vector encoding CRISPR-Cas9 and guide RNA—via subretinal injection to 14 adults. By May 2024, 11 participants (79%) showed gains in best-corrected visual acuity, mobility, and vision-related quality of life, with editing confirmed in photoreceptors and no dose-limiting toxicities or therapy-attributed serious adverse events. Intellia Therapeutics' NTLA-2001 (nexiguran ziclumeran) marked the first systemic application, focusing on transthyretin (TTR) amyloidosis via LNP-mediated CRISPR-Cas9 knockout of the TTR gene in liver cells. The phase 1 trial, initiated in 2021, dosed 31 patients across cardiomyopathy (ATTR-CM) and polyneuropathy (ATTRv-PN) cohorts, achieving mean serum TTR reductions of 93% at one month and durability beyond 24 months in most cases. September 2025 updates from the ATTRv-PN arm revealed 72% of 18 assessed patients with neurological improvements or stabilization per modified Neuropathy Impairment Score, alongside 87% TTR knockdown persistence, prompting phase 3 enrollment in MAGNITUDE-2. In May 2025, a bespoke in vivo CRISPR therapy became the first personalized treatment for an infant with a rare urea cycle disorder stemming from a unique CPS1 mutation. Collaborators including the and NIH designed and manufactured the LNP-delivered editor in six months, administering initial low-dose intravenous infusions starting February 2025. Post-treatment assessments showed biochemical stabilization, symptom amelioration, and medication reductions, demonstrating feasibility for "n-of-1" interventions in ultra-rare conditions while establishing accelerated regulatory precedents. These milestones underscore in vivo CRISPR's potential for one-time cures in monogenic diseases, though hurdles persist: hepatic tropism limits non-liver targets, Cas immunogenicity necessitates protein shielding or transient expression, and long-term oncogenic risks from double-strand breaks drive adoption of nickase-based or prime editing refinements in preclinical pipelines.

Enhanced Imaging and Real-Time Monitoring

Advancements in optical imaging modalities have significantly improved the depth and resolution of in vivo visualizations, enabling real-time tracking of cellular and molecular dynamics in living animal models. Near-infrared window II (NIR-II) fluorescence imaging, which operates in the 1000-1700 nm range, offers enhanced tissue penetration up to several millimeters with minimal scattering and autofluorescence compared to visible or NIR-I wavelengths, facilitating longitudinal studies of tumor progression and drug distribution in mice. This technique has been applied in post-2020 experiments to monitor nanoparticle biodistribution in real time, revealing pharmacokinetic behaviors unattainable with traditional methods. Hybrid imaging systems combining photoacoustic (PA) and fluorescence modalities have emerged as powerful tools for non-invasive, high spatiotemporal resolution monitoring, particularly in neuroscience. A 2025 study demonstrated a light-induced thermoacoustic holographic microscope (LiTA-HM) capable of cortex-wide imaging of neurovascular coupling in rodents, achieving sub-micron lateral resolution over fields of view exceeding 5 mm² with stability for hours-long sessions. Such systems leverage PA's acoustic wave propagation for deeper penetration (up to centimeters) while integrating fluorescence for molecular specificity, outperforming standalone optical approaches in capturing dynamic events like blood flow changes during stimuli. Nanoprobe-based technologies have further refined intracellular real-time imaging, with metallic and organic nanoprobes enabling multiplexed detection of biomarkers in vivo via computed tomography or magnetic resonance enhancements. Clinical translation potential was highlighted in 2025 reviews, where these probes supported sensitive tracking of macrophage infiltration in tumors, quantifying densities with <10% variability in mouse models. Integration of AI algorithms for image processing has accelerated analysis, reducing computational times from hours to seconds and improving predictive accuracy for disease states by 20-30% in high-content screening datasets. Implantable photonic and biosensor arrays have advanced organ-level real-time monitoring, with flexible strain, pressure, and temperature sensors deployed in rodent viscera to capture physiological responses at 1-10 Hz sampling rates. These devices, refined since 2020, provide wireless data transmission over distances up to 10 cm, enabling chronic studies of cardiac or neural function without tethering artifacts. Despite these gains, challenges persist in biocompatibility and signal-to-noise ratios for long-term (>30 days) deployments.

References

  1. [1]
    In vivo vs. in vitro: What is the difference? - MedicalNewsToday
    Aug 30, 2020 · In vivo is Latin for “within the living.” It refers to tests, experiments, and procedures that researchers perform in or on a whole living ...
  2. [2]
    Definition of in vivo - NCI Dictionary of Cancer Terms
    (in VEE-voh) In the body. The opposite of in vitro (outside the body or in the laboratory). Search NCI's Dictionary of Cancer Terms
  3. [3]
    In Vivo vs. In Vitro: Definition, Examples, and More - Healthline
    Aug 19, 2019 · In vivo refers to when research or work is done with or within an entire, living organism. Examples can include studies in animal models or human clinical ...
  4. [4]
    In Vivo vs. In Vitro: What Are the Differences? - Verywell Health
    "In vivo" means research done on a living organism, while "in vitro" means research done in a laboratory dish or test tube.
  5. [5]
    In Vivo vs In Vitro: Definition, Pros and Cons | Technology Networks
    Dec 18, 2023 · In this article, we will define the terms in vivo and in vitro and further evaluate the pros and cons of analysis using these methods with ...<|separator|>
  6. [6]
    Why Are in Vivo Studies Important? | in Vivo Research Benefits
    Sep 16, 2021 · In vivo studies, using live animals, are important for drug development, understanding disease, and informing clinical studies, and are crucial ...
  7. [7]
    [PDF] What are In vivo, In vitro, and In silico Studies?
    In vivo means in a complete living animal. Studies in both humans and animals are called in vivo studies. In vivo studies are usually done after in vitro ...
  8. [8]
    In vitro vs. In vivo: Is One Better? - UHN Research
    Nov 28, 2019 · In vitro and in vivo studies have their merits and flaws, but both are required to holistically evaluate the safety and efficacy of an experimental drug.
  9. [9]
    In Vivo Research Strategies that Reduce Risk of Failure in the Clinic
    Oct 17, 2024 · Maximizing the set-up of early preclinical in vivo research could reduce later failure in the clinic. In addition to providing critical ...
  10. [10]
    In Vivo Imaging in Pharmaceutical Development and Its Impact on ...
    In vivo imaging is recognized as a useful method to provide biomarkers for target engagement, treatment response, safety, and mechanism of action. Imaging ...
  11. [11]
    The Role of in-vivo Studies in Rare Disease Therapeutic Development
    Jul 6, 2023 · In-vivo studies use animal models to test drug efficacy, biodistribution, and toxicity, revealing if a treatment works in a living organism.
  12. [12]
    From in vitro Experiments to in vivo and Clinical Studies - PubMed
    The disadvantage of the in vitro procedures is that they are mostly performed on cancerous cell lines that have a substantially abnormal function. Furthermore, ...
  13. [13]
    [PDF] The role of in vivo imaging in drug discovery and development
    In vivo imaging enables quick drug efficacy screening, provides real-time assessments, improves clinical success, and reduces drug discovery time and costs.
  14. [14]
    In vitro or In vivo. What is the difference? - AINIA
    Sep 5, 2025 · The term “in vivo” comes from Latin and means “within the living.” It refers to experimental studies conducted on whole living organisms, such ...
  15. [15]
    In Vivo Study - an overview | ScienceDirect Topics
    In vivo (Latin meaning within the living) represents a group of experiments performed in living organisms. In an in vivo study, drug molecule/s free or ...Missing: etymology origin
  16. [16]
    In vivo, in vitro and in silico: an open space for the development of ...
    The main consequence of the shift from in vivo/in vitro to in silico research is that a significant fraction of the community has turned inward, with its own ...
  17. [17]
    An Overview of In Vitro, In Vivo, and Computational Techniques for ...
    Dec 10, 2020 · In this paper, we review the standard models for the study of tumor angiogenesis on the macroscopic scale that include in vitro, in vivo, and computational ...
  18. [18]
    Differences between in vitro, in vivo, and in silico studies - the MPKB
    In vivo studies​​ In vivo (Latin for “within the living”) refers to experimentation using a whole, living organism as opposed to a partial or dead organism. ...
  19. [19]
    In Vitro vs. In Vivo vs. In Silico: Preclinical Assay Differences
    Mar 16, 2022 · As they occur within the organism, the results of in vivo studies are considered more reliable or more relevant than those of in vitro studies.
  20. [20]
    An overview of in vitro, ex vivo and in vivo models for studying the ...
    Ex vivo models are considered to be a compromise between in vitro and in vivo models. One major advantage of using functionally isolated tissues as part of an ...
  21. [21]
    (PDF) What is the difference between Ex vivo, In vivo and In vitro
    Oct 14, 2024 · In vitro (Latin: in glass) studies in experimental biology are those that are conducted using components of an.
  22. [22]
    François Magendie (1783–1855) and his contributions to the ...
    François Magendie made epoch-making contributions to experimental physiology, pharmacology, anatomy, and pathology. His name lives on today in the eponyms Bell- ...
  23. [23]
    François Magendie (1783-1855) and his contributions to ... - PubMed
    Additionally, he accurately described the functions of the dorsal and ventral rootlets using vivisection, and realized that the exposed meninges were ...<|separator|>
  24. [24]
    François Magendie: Early French Physiologist
    In 1808, he received the M.D. degree in Paris. Magendie pioneered in experimental physiology, and his work in 1809 marked the beginning of modern pharmacology.
  25. [25]
    Claude Bernard, the Founder of Modern Medicine - PMC - NIH
    May 20, 2022 · His main contribution to physiology and medicine is probably the creation of the concept of «milieu intérieur» and its constancy, which was ...
  26. [26]
    Claude Bernard and life in the laboratory - PMC - NIH
    As already said, Bernard founded his way of practicing physiology on experimentation in vivo. It was clear for him that physiology had to use the means and ...
  27. [27]
    Claude Bernard: bicentenary of birth and his main contributions to ...
    Bernard's main scientific achievements in neurophysiology. Bernard shed light on the role of the pancreas in digestion, and the liver in creating glucose, for ...
  28. [28]
    Claude Bernard, the first systems biologist, and the future of ...
    Dec 18, 2007 · The first systems analysis of the functioning of an organism was Claude Bernard's concept of the constancy of the internal environment.
  29. [29]
    Pavlov and integrative physiology - PubMed - NIH
    Ivan Petrovich Pavlov was the first physiologist to win the Nobel Prize. The Prize was given in 1904 for his research on the neural control of salivary, gastric ...Missing: vivo Sherrington
  30. [30]
    The early history of the synapse: from Plato to Sherrington - PubMed
    One hundred years ago, in 1897, Sherrington adopted the name synapse. However, the concept of the synapse emerged from considerations of how muscles are ...
  31. [31]
    Has NIH Lost Its Halo? - Issues in Science and Technology
    When the NIH budget was $700,000 going into World War II, it was easy to quadruple the budget to $3.4 million by the war's end, and to boost it another tenfold ...
  32. [32]
    [PDF] WORLD WAR II AND THE 20TH-CENTURY EXPANSION OF ...
    WWII's CMR laid the foundation for postwar biomedical innovation, fueled drug development, and shaped the modern US biomedical system, with effects lasting ...
  33. [33]
    3 A HISTORY OF ANIMAL MODELS - MIT Press Direct
    This changed after World War II, when scientists exposed enormous numbers of mice to ionizing radia- tion (Russell, 2013), which greatly increased the animals' ...<|control11|><|separator|>
  34. [34]
    The Era of Biomedicine: Science, Medicine, and Public Health in ...
    Biomedicine, "invented" after WWII, saw increased research, state investment, and a new relationship between science, technology, and medicine, with a new "way ...
  35. [35]
    Clinical trials and medical experiments - Science Museum
    Jul 30, 2019 · The modern definition of informed consent came out of the Nuremberg trials, a series of legal trials between 1945 and 1947 to prosecute ...Missing: milestones vivo post-
  36. [36]
    Historical Overview - Meeting the Nation's Needs for Biomedical and ...
    Federal interest and involvement in biomedical and behavioral research increased dramatically after World War II largely as a result of the demonstration during ...
  37. [37]
    The Importance of Murine Models in Determining In Vivo ... - MDPI
    This review describes the role of murine models in the drug development pipeline. It also explains some in vivo pharmacokinetic, safety, and efficacy ...
  38. [38]
    Role of animal models in biomedical research: a review - PMC
    Jul 1, 2022 · In recent advancements, animal models are the most practical tools for pre-clinical drug screening before application into clinical trials.
  39. [39]
    The Role of Mouse Models in Drug Discovery | Taconic Biosciences
    Mar 20, 2025 · Discover how mouse models drive drug discovery, from disease modeling to preclinical testing, enhancing research efficiency and accelerating ...
  40. [40]
    An Updated Comparison of Common Model Organisms
    In this blog, we provide an overview of many commonly used model organisms, and compare their advantages and limitations.Introduction · Caenorhabditis Elegans... · Danio Rerio (zebrafish)
  41. [41]
    A Model of Discovery: The Role of Imaging Established ... - Frontiers
    This review seeks to synthesize current knowledge of established and emerging non-mammalian model organisms with advances in cellular-resolution in vivo imaging ...
  42. [42]
    A large-scale in vivo screen to investigate the roles of human genes ...
    Oct 7, 2024 · Genetic screens using Drosophila are a useful way of testing large number of genes to study their function and roles within signaling pathways.
  43. [43]
    Drosophila melanogaster: A Model Organism to Study ... - Frontiers
    The fruit fly, Drosophila melanogaster, is used as a model organism to study disciplines ranging from fundamental genetics to the development of tissues and ...
  44. [44]
    Animal models in biomedical research: Relevance of Drosophila ...
    Jan 15, 2025 · Drosophila melanogaster is becoming increasingly relevant as preferred model for understanding the biochemical basis of several human diseases.
  45. [45]
    Caenorhabditis elegans as a model system for studying aging ...
    C. elegans ND models serve as powerful tools for studying the molecular pathology governing aggregate formation and disease progression.
  46. [46]
    In-Vivo Quantitative Image Analysis of Age-Related Morphological ...
    Jun 19, 2019 · We found that, during aging, the soma of neurons decreases in volume, whereas the number and length of neurite outgrowths from the soma both increase.
  47. [47]
    An automated compound screening for anti-aging effects on ... - Nature
    Aug 24, 2017 · C. elegans with its powerful genetics and a well-defined nervous system has become a prominent model organism for studying in vivo the process ...
  48. [48]
    In vivo drug discovery in the zebrafish - Nature
    Jan 1, 2005 · In this article, we will review the use of zebrafish in target validation, disease modelling, target and lead compound discovery, and toxicology.
  49. [49]
    The zebrafish embryo as an in vivo model for screening nanoparticle ...
    Jan 26, 2022 · We propose the zebrafish embryo TB-model as a quick and sensitive tool for evaluating the in vivo toxicity and efficacy of new anti-TB compounds.Results · In Vivo Toxicity Assays In... · Zebrafish Survival Study
  50. [50]
    Diving into drug-screening: zebrafish embryos as an in vivo platform ...
    Consequently, zebrafish embryos present a valuable tool for predicting antibacterial compound activity and mitigating early failures in drug research and ...Introduction · Regulatory considerations in... · Drug-screening strategies in...
  51. [51]
    Towards zebrafish model applications in drug discovery targeting ...
    Sep 5, 2025 · This review provides a comprehensive overview of zebrafish applications in CNS drug discovery and neurotoxicity assessment.
  52. [52]
    Use of animals for toxicology testing is necessary to ensure patient ...
    Commentary. Use of animals for toxicology testing is necessary to ensure patient safety in pharmaceutical development.
  53. [53]
    The Crucial Role of preclinical toxicology studies in Drug Discovery
    The safety and efficacy assessments that in vivo toxicology studies provide include: Early Safety Assessment: To assess the safety and toxicity profile of ...
  54. [54]
    Toxicology Studies - Pacific BioLabs
    In Vivo Toxicology Test Categories · Acute Systemic Toxicity (3 to 14 days) · Subacute Toxicity (14 to 30 days) · Subchronic Toxicity (longer than 30 days) · IND ...
  55. [55]
    6 Types of Toxicology Studies for IND & NDA Programs - WuXi AppTec
    May 2, 2024 · #1. General Toxicology Testing · #2. Carcinogenicity Testing · #3. Safety Pharmacology Testing · #4. DART Testing · #5. In Vitro Toxicology Testing.
  56. [56]
    In Vivo Toxicity Study - Creative Bioarray
    The main specific contents of the study are ① acute toxicity test, ② chronic or sub-chronic toxicity test, ③ genetic toxicity test, ④ reproductive toxicity test ...
  57. [57]
    [PDF] Roadmap to Reducing Animal Testing in Preclinical Safety Studies
    Apr 10, 2025 · This roadmap outlines a strategic, stepwise approach for FDA to reduce animal testing in preclinical safety.
  58. [58]
    FDA Announces Plan to Phase Out Animal Testing Requirement for ...
    Apr 10, 2025 · ... toxicity and cell lines and organoid toxicity testing in a laboratory setting (so-called New Approach Methodologies or NAMs data).
  59. [59]
    The (misleading) role of animal models in drug development
    Apr 7, 2024 · Animals like mice and rats have long been used in medical research to help understand disease and test potential new treatments before human ...Introduction · How and why are animal... · What are alternatives to animal...
  60. [60]
    The role of early in vivo toxicity testing in drug discovery toxicology
    The role of early in vivo toxicity testing in drug discovery toxicology ... Models, Animal*; Predictive Value of Tests; Species Specificity; Time Factors ...
  61. [61]
    The evolving role of investigative toxicology in the pharmaceutical ...
    Feb 13, 2023 · Investigative toxicology can be broadly split into two approaches: the prediction of potential toxicity before a compound is tested non- ...
  62. [62]
    Five Popular Model Organisms - Addgene Blog
    Apr 11, 2019 · The five popular model organisms are: house mouse, fruit fly, yeast, zebrafish, and the worm.Missing: review | Show results with:review
  63. [63]
    Small Model Organisms as Versatile Research Tools | The Scientist
    Jul 26, 2024 · Flies, worms, and fish with rapid development and characterized genetics help scientists uncover fundamental and universal biological processes.
  64. [64]
    In Vivo RNAi-Based Screens: Studies in Model Organisms - PMC
    Nov 25, 2013 · In this review, we focus mainly on organism-based (hereafter referred to as in vivo) RNAi screens and experimental and computational ...
  65. [65]
    Overview: Generation of Gene Knockout Mice - PMC - NIH
    The knockout mouse has been a valuable tool for geneticists to discern the role of a gene in embryonic development and in normal physiological homeostasis. Mice ...
  66. [66]
    Knockout Mice Fact Sheet
    Aug 17, 2020 · About 15 percent of gene knockouts are developmentally lethal, which means that the genetically altered embryos cannot grow into adult mice. The ...
  67. [67]
    Developmental studies of Brca1 and Brca2 knock-out mice - PubMed
    Brca1 and Brca2 knock-out mice show embryonic lethality and a cellular proliferation defect, associated with p53 pathway activation.
  68. [68]
    Analyses of Conditional Knockout Mice for Pogz, a Gene ... - MDPI
    We created two lines of conditional knockout mice for Pogz, one specific to excitatory neurons (Emx1-Pogz mice) and the other to inhibitory neurons (Gad2-Pogz ...
  69. [69]
    Revolutionizing in vivo therapy with CRISPR/Cas genome editing
    CRISPR/Cas9 technology has enabled scientists to make precise changes to the DNA of living organisms, including humans, allowing for the insertion, deletion, or ...
  70. [70]
    CRISPR/Cas9 therapeutics: progress and prospects - Nature
    Jan 16, 2023 · CRISPR/ Cas9 is a highly effective gene-editing tool that is widely used in the scientific community. The CRISPR/Cas9 system evolved naturally ...
  71. [71]
    In vivo genome editing via CRISPR/Cas9-mediated homology ...
    May 6, 2024 · Here, we apply CRISPR/Cas9 mediated homology-independent targeted integration (HITI)-based gene editing therapy in HEK293T cells, BCD patient derived iPSCs, ...
  72. [72]
    Estrogen receptor (ER)β knockout mice reveal a role for ERβ ... - PNAS
    We now show that ERβ is necessary for late embryonic development of the brain and is involved in both neuronal migration and apoptosis.Sign Up For Pnas Alerts · Brdurd Labeling And Analysis · Results
  73. [73]
    Quantitative In Vivo Imaging of Embryonic Development
    Advances in in vivo imaging have transformed the study of embryonic development and contributed many new insights into dynamic processes of tissue and organ ...
  74. [74]
    Genetic Background Matters: Population-Based Studies in Model ...
    Jul 8, 2022 · In this review, we describe many panels of organisms and examples of how modeling diseases on them can accelerate the pace of discoveries ...
  75. [75]
    Model organisms contribute to diagnosis and discovery in the ...
    May 7, 2021 · Platforms employing model organisms enable discovery of novel gene-disease relationships, help establish variant pathogenicity, and often lead ...
  76. [76]
    Animal models in the drug discovery pipeline for Alzheimer's disease
    This review paper summarizes and critically evaluates currently available animal models, and discusses their value to the Alzheimer drug discovery pipeline.Amyloid-β Infusion Rodent... · Transgenic Rodent Models For... · Table 1
  77. [77]
    In vitro and in vivo drug screens of tumor cells identify novel ...
    Dec 20, 2021 · These data indicate that in vitro and in vivo drug screening of tumor cells could increase therapeutic options and improve clinical outcomes for high‐risk ...Patients And Tumor Samples · In Vitro Drug Screening... · Validation Of Tumor Cell...
  78. [78]
    Experimental modelling of Alzheimer's disease for therapeutic ...
    This review aims to summarise the experimental models employed for AD at present, and discuss their role in the drug development process.8. In Vitro Models Of Ad · 9. In Vivo Models Of Ad · 9.1. Therapeutics Tested In...
  79. [79]
    Why 90% of clinical drug development fails and how to improve it?
    If drug candidates in the preclinical stage are also counted, the failure rate of drug discovery/development is even higher than 90%. Analyses of clinical trial ...
  80. [80]
    Preclinical Development: The Safety Hurdle Prior to Human Trials
    Apr 30, 2016 · Preclinical testing is the final deciding gate prior to clinical studies, and only 12% of the candidates advance to the Phase I clinical trial.
  81. [81]
    Animal models in translational medicine: Validation and prediction
    In this review, the current challenges and limitations of animal models are discussed, with a focus on the fit-for-purpose validation.
  82. [82]
    Understanding Validity in Animal Models for Drug Discovery
    Dec 20, 2024 · Animal model validity includes predictive (predicting outcomes), face (replicating human disease), and construct (reflecting disease mechanisms ...
  83. [83]
  84. [84]
    Live Cell in Vitro and in Vivo Imaging Applications - NIH
    Quantitative in vitro and in vivo imaging represents a more holistic approach to evaluation of drug efficacy, providing an unbiased and thus more opportunistic ...
  85. [85]
    In Vivo vs. In Vitro: Advantages & Disadvantages - MedicineNet
    In vivo studies are "within the living" and more reliable, while in vitro studies are "in the glass" and done in labs, often on cell cultures.
  86. [86]
    Advancing predictive toxicology: overcoming hurdles and shaping ...
    Jan 6, 2025 · In vivo analysis tests a compound's effect within a living system, encompassing the full spectrum of biological interactions such as absorption, ...In Silico Toxicology And The... · Data Availability And... · In Vivo Relevance
  87. [87]
    [PDF] Quantitative variability in repeat dose toxicity studies - EPA
    Dec 17, 2019 · In silico, in chemico, and in vitro models cannot predict in vivo systemic effect values with greater accuracy than those animal models ...<|separator|>
  88. [88]
    Animal to human translation: a systematic scoping review of ...
    Jul 15, 2019 · The concordance rates between animal and human toxicity were 71% when all species were considered, with nonrodents alone predictive for 63% and ...
  89. [89]
    Acute Systemic Toxicity: Alternative in Vivo and in Vitro Methods
    Currently, there is none of the in vitro alternative methods or their combinations which could fully replace in vivo methods for acute systemic toxicity.
  90. [90]
    On In Vivo vs. In Vitro Models
    May 23, 2024 · In vivo models also allow for the evaluation of long-term effects, chronic conditions, and the overall impact on the organism's physiology.
  91. [91]
    Pre-clinical animal models are poor predictors of human toxicities in ...
    Sep 1, 2020 · Their analysis showed that a combination of rodent and non-rodent studies had a positive concordance for values in humans for 71% of drugs, and ...<|control11|><|separator|>
  92. [92]
    Advancing Toxicity Predictions: A Review on in Vitro to in Vivo ... - NIH
    May 9, 2024 · As a key step in next-generation risk assessment (NGRA), in vitro to in vivo extrapolation (IVIVE) aims to mobilize a mechanism-based understanding of ...
  93. [93]
    Ethical considerations regarding animal experimentation - PMC - NIH
    Due to the ethical issues surrounding the use of animals in experimentation, their use is declining in those research areas where alternative in vitro or in ...
  94. [94]
    The dark side of the animal experiments - PMC - PubMed Central
    Jul 6, 2022 · Finally, most of the animals in experiments may suffer from unnecessary pain and stress beyond the aim of experiment. Exposure of animals to ...
  95. [95]
    Facts and figures on animal testing | Cruelty Free International
    Of the 9.3 million uses of animals in 2022, 49% were considered by researchers to have caused moderate or severe suffering to the animals involved.
  96. [96]
    The Flaws and Human Harms of Animal Experimentation - PMC - NIH
    Nonhuman animal (“animal”) experimentation is typically defended by arguments that it is reliable, that animals provide sufficiently good models of human ...
  97. [97]
    Problems with Animal Research - American Anti-Vivisection Society
    AAVS believes that animals have the right to not be exploited for science, and that we should not have to choose between helping humans and harming animals.Missing: vivo | Show results with:vivo
  98. [98]
    The continued importance of animals in biomedical research - Nature
    Oct 14, 2024 · Animal models provide a comprehensive, whole-organism perspective that alternative methods cannot yet fully replicate.
  99. [99]
    The ethics of animal research. Talking Point on the use of animals in ...
    No responsible scientist wants to use animals or cause them unnecessary suffering if it can be avoided, and therefore scientists accept controls on the use of ...Missing: vivo | Show results with:vivo
  100. [100]
    Inter-species differences in drug properties - PubMed
    There has been a clear trend towards decreased reliance upon animal studies and increased emphasis upon experiments with human-derived tissues.
  101. [101]
    Inter-species differences between humans and other mammals in ...
    This study investigated the metabolic transformation of carbofuran in seven species of mammals using LC-MS/MS and liver microsomes.
  102. [102]
    Limitations of Animal Studies for Predicting Toxicity in Clinical Trials
    Nov 25, 2019 · Percentages of drugs that fail in preclinical trials (due to drug toxicity or failure of efficacy in animal testing) and in clinical trials (due ...
  103. [103]
    Replacing mammals in drug development: Non‐mammalian models ...
    Nov 24, 2022 · Driven by economics and pressure to reduce the number of mammals, non‐mammalian animal models are becoming valuable alternatives for translational and ...
  104. [104]
    5% of animal studies lead to human treatments – failure or success?
    Jun 24, 2024 · Uncover the debate on animal research success rates - only 5% lead to human treatments. Is it failure or a step in the right direction?
  105. [105]
    Using Zebrafish for High-Throughput Screening of Novel ... - JACC
    The complexity of the underlying biology is a major contributor to the current failure rate in drug discovery. For example, drugs that target the ...
  106. [106]
    Research on the Species Difference of the Hepatotoxicity ... - Frontiers
    Apr 22, 2021 · Understanding the differences in drug toxicity among species can help better interpret results from in vivo studies, lead to more precise ...
  107. [107]
    Poor Translatability of Biomedical Research Using Animals - PubMed
    Mar 7, 2023 · The failure rate for the translation of drugs from animal testing to ... animal models has failed to offer effective new treatments. We ...Missing: interspecies gaps
  108. [108]
    Statistical analysis of preclinical inter-species concordance of ...
    For example, liver toxicity concordance in short-term studies between female rats and dogs observed an average LR+ of 1.84, and an average LR- of 0.73. This was ...
  109. [109]
    Human disease models in drug development - Nature
    May 11, 2023 · In this Review, we discuss preclinical and clinical studies that benefited from these models, focusing on organoids, bioengineered tissue models and organs-on- ...
  110. [110]
    Costs of Animal and Non-Animal Testing - Humane World for Animals
    Oct 23, 2012 · Costs of Animal and Non-Animal Testing ; Rat developmental toxicity test, animal test, $50,000 ; Rat limb bud test, in vitro test, $15,000 ; Non- ...
  111. [111]
    In vivo Base Study Rates at the College of Pharmacy at HSC.
    May 6, 2025 · In vivo Base Study Rates ; Pharmacokinetics, Rat, $1,100 ; Pharmacokinetics, Mouse, $257 ; Neonatal Sepsis, Rat, $463 ; Intrauterine, Mouse, $592.
  112. [112]
    [PDF] RccHan®:WIST rat in toxicology studies - Inotiv
    The per-study cost estimate (in-life study costs only) between these strains ... Per Animal Cost. $47.60. $46.50. $51.20. $41.25. Total Animal Cost. $7,904.
  113. [113]
    [PDF] Wasted Money in United States Biomedical and Agricultural Animal ...
    Academic "administrative costs" consume one in three research grant dollars, approximately us$9.3 billion ( one-fourth) of the. Kathrin Herrmann and Kimberley ...
  114. [114]
    Step 2: Preclinical Research - FDA
    Jan 4, 2018 · These regulations set the minimum basic requirements for: study conduct. personnel. facilities. equipment. written protocols. operating ...
  115. [115]
    In Vivo Services (IVS) Core - UW Research
    All services through the IVS Core are charged on an hourly tech time rate. Type: Service. Rodent colony management, in vivo experimental study execution, in ...
  116. [116]
    Guidelines for planning and conducting high-quality research and ...
    Jul 10, 2020 · This includes, among other things, their education and training, personal protection, their workload, and means of ensuring adequate staffing ...<|control11|><|separator|>
  117. [117]
    In Vitro Models: An Alternative to In Vivo Studies - Visikol
    Aug 24, 2023 · The scalability of in vitro models may vary depending on the specific model being used, but they can generally be adapted to high-throughput ...
  118. [118]
    What Do Experimental Mice Really Cost? - The Jackson Laboratory
    Mar 6, 2024 · This framework is designed to trim costs, save time, and reduce the number of animals used for colony maintenance and cohort production.
  119. [119]
    Limitations of Animal Studies for Predicting Toxicity in Clinical Trials
    Animal testing is used in pharmaceutical and industrial research to predict human toxicity, and yet analysis suggests that animal models are poor predictors of ...
  120. [120]
    The 3Rs - NC3Rs
    The 3Rs were first defined by Russell and Burch in their book The Principles of Humane Experimental Technique . The NC3Rs has updated the definitions in line ...Missing: origin | Show results with:origin
  121. [121]
    Russell and Burch's 3Rs Then and Now: The Need for Clarity in ...
    WMS Russell (1925–2006) and RL Burch (1926–1996) originated the concepts of replacement, reduction, and refinement, which they published in their 1959 book, The ...
  122. [122]
    Animal Use Alternatives (3Rs) | National Agricultural Library - USDA
    The 3Rs alternatives refers to the replacement, reduction, and refinement of animals used in research, teaching, testing, and exhibition.
  123. [123]
    Strategies to apply 3Rs in preclinical testing - PMC - PubMed Central
    Oct 5, 2021 · This article describes different approaches to apply 3Rs in preclinical experimentation for either research or regulatory purposes.
  124. [124]
    Creative Implementation of 3Rs Principles within Industry Programs
    First and foremost in the United States are the responsibilities of the IACUC to apply the principles of the 3Rs to every aspect of animal experimentation.
  125. [125]
    Ethical use of animals in medicine testing
    3Rs principles · replacing the use of animals with non-animal methods where possible; · reducing the number of animals used to a minimum while still obtaining ...
  126. [126]
    Regulatory Applications of 3Rs - National Toxicology Program - NIH
    Jun 30, 2025 · This page provides examples of how US federal agencies have applied 3Rs approaches to testing requirements.
  127. [127]
    The IACUC | OLAW - NIH
    Oct 30, 2024 · The IACUC is responsible for oversight of the animal care and use program and its components as described in the Public Health Service (PHS) Policy.Missing: vivo | Show results with:vivo
  128. [128]
    The Role of IACUCs in Responsible Animal Research - PMC
    Nov 11, 2019 · The central role of IACUCs in research animal use and oversight helps it safeguard responsible animal research by ensuring ethical, scientifically sound, ...
  129. [129]
    Regulation of Animal Research - NCBI - NIH
    The first federal law regulating animal research was the Laboratory Animal Welfare Act passed by Congress in 1966.
  130. [130]
    Federal Laws and Agencies Involved With Animal Testing
    The United States Department of Agriculture, or USDA, is the federal agency responsible for overseeing and inspecting laboratories that experiment on animals, ...Missing: vivo | Show results with:vivo
  131. [131]
    U.S. Government Principles for the Utilization and Care of Vertebrate ...
    Jul 30, 2025 · They provide the ethical framework for currently accepted approaches in the care and housing of research animals.
  132. [132]
    FDA Roadmap to Reducing Animal Testing: A New Regulatory Era
    Jul 8, 2025 · This initiative builds on the 2022 FDA Modernization Act 2.0, which legally removed the requirement for animal testing in IND and BLA ...
  133. [133]
    Directive - 2010/63 - EN - EUR-Lex - European Union
    Directive 2010/63/EU of the European Parliament and of the Council of 22 September 2010 on the protection of animals used for scientific purposes.
  134. [134]
    Protection of laboratory animals | EUR-Lex - European Union
    WHAT IS THE AIM OF THE DIRECTIVE? Directive 2010/63/EU sets out measures designed to protect animals used for scientific purposes, including animals used in ...
  135. [135]
    European Union Legislation for the Welfare of Animals Used for ...
    Jul 20, 2023 · The Directive 2010/63/EU currently provides a high welfare standard for animals used for scientific purposes. Furthermore, the amendment of the ...Missing: testing | Show results with:testing
  136. [136]
    Oversight :: NABR.ORG
    A comprehensive system of government oversight is in place to regulate the use of animals in scientific research.
  137. [137]
    Unified ethical principles and an animal research 'Helsinki ...
    The EU Directive ensures that for all animal research there is a designated veterinarian and regulatory animal welfare oversight body. It guides the conduction ...
  138. [138]
    Gene Editing for CEP290-Associated Retinal Degeneration
    May 6, 2024 · In preclinical models, treatment with EDIT-101 resulted in sustained CEP290 gene editing at levels that met the therapeutic threshold. The ...
  139. [139]
    Participants of pioneering CRISPR gene editing trial see vision ...
    May 6, 2024 · The BRILLIANCE trial results showed 11 out of 14 treated participants experienced some improvements in vision and quality of life measures.
  140. [140]
    CRISPR-Cas9 In Vivo Gene Editing for Transthyretin Amyloidosis
    Jun 26, 2021 · NTLA-2001 is an in vivo gene-editing therapeutic agent that is designed to treat ATTR amyloidosis by reducing the concentration of TTR in serum.
  141. [141]
    Nexiguran Ziclumeran Gene Editing in Hereditary ATTR with ...
    Sep 25, 2025 · The results support further clinical investigation in a phase 3 trial (MAGNITUDE-2; NCT06672237). Notes. This article was published on September ...
  142. [142]
    Release Details - Intellia Therapeutics - investor relations
    Sep 25, 2025 · Intellia began dosing patients in the Phase 3 MAGNITUDE-2 trial in April 2025 . Patient screening is advancing rapidly, and enrollment ...
  143. [143]
    Infant with rare, incurable disease is first to successfully ... - NIH
    May 15, 2025 · Thursday, May 15, 2025. Infant with rare, incurable disease is first to successfully receive personalized gene therapy treatment. NIH ...
  144. [144]
    Patient-Specific In Vivo Gene Editing to Treat a Rare Genetic Disease
    May 15, 2025 · We developed a workflow for the rapid development of customized, corrective gene-editing therapies for patients with ultrarare or unique “N-of-1” variants.
  145. [145]
    First Patient Treated with On-Demand CRISPR in Just 6 Months
    May 15, 2025 · A personalized in vivo CRISPR therapy for an infant was developed and delivered to the patient in just six months.
  146. [146]
    Recent advances in therapeutic gene-editing technologies
    Jun 4, 2025 · This review highlights recent developments of genetic and epigenetic editing tools and explores preclinical innovations poised to advance the field.
  147. [147]
    In vivo delivery of CRISPR-Cas9 therapeutics - PubMed Central - NIH
    Many diseases require safe and efficient in vivo delivery of CRISPR-Cas9 components in order to enable accurate genome editing in target cells and tissues.
  148. [148]
    Advances in the near-infrared Ⅱ for in vivo fluorescence imaging ...
    NIR-II imaging technology spans multiple scales, from macroscopic to microscopic, enabling comprehensive in vivo visualization, becoming a current research ...
  149. [149]
    In Vivo Ultrasound and Photoacoustic Imaging of Nanoparticle ...
    Feb 5, 2025 · Results highlight the advantages of US/PA imaging for cancer immunotherapies, including multiplex imaging of exogenous and endogenous absorbers.
  150. [150]
    Photoacoustic and fluorescence hybrid microscope for cortex-wide ...
    Jul 23, 2025 · In vivo experiments demonstrated the LiTA-HM's capability in cortex-wide NVC imaging in terms of spatiotemporal resolution, FOV, and stability.
  151. [151]
    High-resolution transcranial optical imaging of in vivo neural activity
    Oct 21, 2024 · We show holographic imaging provides a pathway for high-resolution, label-free, non-invasive recording of transcranial in vivo neural activity at depth.
  152. [152]
    Clinical applications of nanoprobes of high-resolution in vivo imaging
    Jan 17, 2025 · Nanoprobes are known for their noninvasive, highly sensitive, real-time IVI and can even be expanded to intracellular imaging.
  153. [153]
    Noninvasive in vivo imaging of macrophages: understanding tumor ...
    Jan 26, 2025 · This study comprehensively reviews several methods for labeling macrophages and various imaging modalities, assessing the merits and drawbacks of each approach.<|separator|>
  154. [154]
    Advances in High-Content Imaging and Informatics: A Joint Special ...
    The clever addition of live-cell imaging to the screening process ensured an easy removal of false positives caused by toxicity. The authors then followed up ...
  155. [155]
    Implantable physical sensors for in vivo organ monitoring | Med-X
    Jan 8, 2025 · This review presents recent progress in four key types of implantable physical sensors: strain sensors, pressure sensors, temperature sensors, and flow sensors.
  156. [156]
    In-vivo continuous monitoring with biosensors based on engineered ...
    The development of biosensors for continuous sensing of biomarkers and drugs, such as small molecule drugs, peptides and proteins such as therapeutic ...