
Type 1 Diabetes Mellitus
ICD-10 Code E10
What is the clinical spectrum of Type 1 Diabetes Mellitus?
Type 1 diabetes mellitus (T1DM) is one of the two main subtypes of the metabolic disease diabetes mellitus (DM). Its etiology, pathophysiology and treatment are drastically different from Type 2 diabetes mellitus (T2DM).
T1DM is an autoimmune disease that typically presents in children or adolescents. It is characterized by chronic hyperglycemia, caused by the T-cell-mediated destruction of pancreatic insulin-producing beta-cells and a subsequent insulin deficiency.
Additional pathophysiological features of T1DM include weight loss and ketoacidosis, due to increased breakdown of fat and muscle, to compensate for the lack of entry of glucose into cells in absence of insulin. The autoimmune attack and the resulting tissue damage leads to chronic inflammation in the pancreas Zajec et al., Genes, Apr 2022, Sapra & Bhandari, StatPearls, Jun 2023, Tomic et al., Nature Rev. Endocrin., Jun 2022, Redondo & Morgan, Nature Rev. Imm., Jun 2023, Patel et al., Biomed. & Pharmaco., Apr 2019.
T1DM is associated with an increased risk of several other diabetic complications, such as cardiovascular disease, nephropathy, retinopathy, diabetic wounds, psychiatric disorders, dental issues, and peripheral neuropathy.
Over half of all limb amputations are due to impaired wound healing, most often triggered by hyperglycemia, chronic inflammation, circulatory dysfunction, hypoxia, autonomic and sensory neuropathy, and impaired neuropeptide signaling.
T1DM exhibits heterogeneous phenotypes, with inter-individual variability in age of onset which can range from childhood to adulthood, in the rate of beta-cells destruction which can be rapid in some individuals and slowly progressive in others, and in the type of immune cells involved in the autoimmune attack.
What do we know about the etiology of Type 1 Diabetes Mellitus?
Much of our understand of mechanisms that underly Type 1 diabetes mellitus (T1DM) is based on animal research, however, animal models of T1DM are poor proxies for studying disease mechanisms of human T1DM.
Genetics plays a major role in the development of pancreatic islet autoimmunity. Genetic factors that increase the risk of developing T1DM include polymorphisms in human leukocyte antigen (HLA) and other immune system-related genes Skyler et al., Diabetes, Dec 2016.
Both endogenous factors, such as dietary content and intestine microbiota, and exogenous factors, such as environmental toxins and viral infections, can trigger the autoimmune attack in genetically susceptible individuals.
Within individuals with T1DM, epidemiological, clinical, genetic, immunological, histological and metabolic differences suggest heterogeneity in pathophysiological mechanisms Sapra & Bhandari, StatPearls, Jun 2023, Zajec et al., Genes, Apr 2022, Redondo & Morgan, Nature Rev. Imm., Jun 2023.
Consequently, T1DM spectrum of endotypes 1 (T1DE1) and 2 (T1DE2) were proposed, according to the age of onset, degree of beta-cells destruction, presence or absence of insulitis, the type of HLA allele, abundance CD8+ T and CD20+ B cells, quality of proinsulin processing and degree of proinsulin to C-peptide ratio.
In over 50 associated loci identified by GWAS and other genetic association studies, the majority of T1DM-associated variants are involved in gene regulation and only HLA-II, INS, and PTPN22 show substantial effects on T1DM.
In both T1DM and T2DM, pathophysiological mechanisms interact in complex ways, leading to the development and progression of DM.
Recently, several shared risk genes or genetic polymorphisms between T1DM and T2DM were found.
Some of the genes located in shared risk loci interact to mutually regulate important pancreatic islet functions. The regulatory impacts on shared genes and pathways generate overlapping biological mechanisms, which mediate pleiotropic effects on T1DM and T2DM.
How similar are human and animal endocrine systems?
This is not an exhaustive list of species-specific differences, nor can one be made given their unknown full extent, but rather an example of how these differences impact the face, construct, predictive, and intrinsic validity of animal models.
Not all species-specific differences can be accounted for in animal models, as there are hundreds of them, their relevance to Type 1 diabetes mellitus (T1DM) is unclear, and their interaction with other organ systems in the animal model makes it difficult to predict how they would have behaved within the human system.
Endocrine systems of human and non-human animals differ at every level, from molecular to organism level, contributing to poor translation of animal studies to humans.
Species-specific differences in structure and function of pancreatic islets
Inter-species differences in cytoarchitecture of pancreatic islets of Langerhans have consequences on the function of islets, susceptibility to diabetes, and disease pathophysiology.
In the islets of Langerhans, alpha, beta, delta, gamma and epsilon cells secrete hormones insulin, glucagon, somatostatin, pancreatic polypeptide and ghrelin, that by regulating, stimulating and inhibiting each other through paracrine and autocrine communication, maintain balanced blood glucose levels and metabolic homeostasis.
In human islets, most beta, alpha, and delta cells are aligned along blood vessels with no particular order, indicating that islet microcirculation likely does not determine the order of paracrine interactions Bosco et al., Diabetes, May 2010.
Imaging of cytoplasmic free Ca2+ concentration, [Ca2+]i, has shown that beta cell oscillatory activity was not coordinated throughout the human islet as it was in mouse islets, suggesting inter-species differences in beta-cell network dynamics and insulin release patterns.
Mathematical modelling of Ca2+ activity in human and mouse islets showed that Ca2+ activity in human islets was more vulnerable to inhibition of highly connected beta cells, termed hubs, than in mouse islets Lei et al., Islets, Aug 2018. In contrast to human islets, mouse islets have a more uniform beta cell distribution, making them less dependent on hubs. This observation suggests that humans may be more susceptible than mice to loss of hubs, that can occur through autoimmune attack (T1DM) or toxicity/inflammation (T2DM).
Furthermore, human islets responded with an increase in [Ca2+]i when lowering the glucose concentration, which could be explained by the fact that human islets contain proportionally fewer beta cells (that produce and secrete insulin, which lowers blood glucose) and more alpha cells (that produce and secrete glucagon, which raises blood glucose) than do mouse islets Cabrera et al., PNAS, Feb 2006, pointing to inter-species differences in glucose regulation.
The neurotransmitter acetylcholine has a major role in the function of the insulin-secreting pancreatic beta cells. Cholinergic input to exocrine acini ducts and endocrine islets of Langerhans stimulates secretion of digestive enzymes and hormones.
In contrast to mouse islets, cholinergic innervation of human islets is sparse. Instead, the alpha cells of human islets provide paracrine cholinergic input that primes the beta cell to respond optimally to increases in glucose concentration. Cholinergic signaling pathways within human islets could potentially be a therapeutic target in diabetes Rodriguez-Diaz et al., Nature Med., Jun 2011. However, because of inter-species differences in pancreatic islets structure and function, this therapeutic lead, and probably many other leads that have the potential to benefit patients, are likely to have failed in animal models of diabetes.
Species-specific differences in gene expression
As it is the case for the overwhelming majority of species, humans have a single insulin gene. Rodents, however, have two non-allelic insulin genes that, despite their differences in structure, are both equally expressed. It is unclear why rodents have two insulin genes and whether they might have different functions, which limits extrapolation to the single insulin gene system in humans Hay & Docherty, Diabetes, Dec 2006, Irwin, Front. Endocrinol., Apr 2021.
The effects of insulin gene duplication in rodents may be further amplified, since insulin transcriptionally regulates the expression of more than 150 genes in various tissues.
Differences in sequences of insulin genes between humans and animals affect the regulation of expression of insulin genes, as well as the structure and function of encoded proteins Chandrasekera & Pippin, Altex, May 2014.
Overall homology of the insulin promoter between humans and rodents is only around 45 to 48%. Many features of cis-regulatory elements and trans-acting factors are differentially regulated in human and mouse islets Hay & Docherty, Diabetes, Dec 2006,
There are also marked inter-species divergences in DNA binding between mouse and human glucose regulatory transcription factors. As much as 41%-89% of the orthologous promoters bound by a protein in one species were not bound by the same protein in the second species.
Species-specific differences in cis regulatory elements (transcription factor binding motifs, epigenetic marks) and trans regulatory elements (expression levels, post-translational modifications) affect how in each species the glucose related-genes respond to insulin.
Subsequently, the auto-regulatory mechanisms by which genes involved in glucose metabolism regulate their own expression and expression of other related genes via cis and trans inputs, diverge between humans and other species.
For instance, while the gene expression of gluconeogenic enzyme phosphoenolpyruvate carboxykinase is highly regulated by insulin in rodents, this insulin-mediated repression is less pronounced in humans, suggesting alternative regulatory pathways Cherrington et al., Endocrinology, Jun 2011.
The inter-species differences in insulin protein sequence and post-translational modifications are likely to manifest in inter-species differences in insulin function and proteolytic processing.
For example, in guinea pigs, that have highly divergent sequences of insulin genes, the biological activities of these insulins differ, acting more as a growth factor than as a metabolic hormone Irwin, Front. Endocrinol., Apr 2021.
Similarly, as a consequence of specific changes in sequences of insulin genes, some species of New World monkeys have insulin hormones with lower potency, despite sharing 85.5% identity with the human insulin precursor.
Of particular relevance for T1DM, inter-species differences in insulin protein sequence can influence how the immune system recognizes insulin and targets insulin-producing beta cells. In case of T2DM, they may impact how insulin binds to its receptor and thereby produce inter-species divergences in pathophysiology of insulin resistance. The response of T1DM animal models in preclinical trials to gene therapies via introduction the human insulin gene may yield results that are not translatable to humans.
Species-specific differences in glucose regulation
Even though insulin is essential for maintaining optimum blood glucose levels in all vertebrate species, its function has evolved in species-specific manners.
The two most frequently used species to model T1DM/T2DM, mice and rats, differ significantly from humans at gene, protein, pathway, cellular, tissue, organ and organism levels of glucose regulation Chandrasekera & Pippin, Altex, May 2014.
Notably, inter-species differences between rodents and humans were showed in intracellular signal transduction and glucose metabolic pathways.
For example, glucose metabolism via anaplerotic enzymes pyruvate carboxylase and ATP citrate lyase was reported to be lower in human islets compared to rodent islets. Compared to rodent islets, human islets appeared to be less dependent on PC for insulin secretion MacDonald et al., J. Biol. Chem., Mar 2011. Such differences in glucose metabolic pathways are critical for understanding the regulation of human glucose homeostasis under normal and diabetic conditions.
The principal glucose transporter present in rodent beta cells is glucose transporter 2 (GLUT2), and greatly reduced GLUT2 expression levels have been shown to correlate with elements of T2DM in various diabetic rodent models. However, human islets predominantly express GLUT1 and GLUT3, and GLUT2 expression levels do not correlate with human T2DM van den Bunt & Gloyn, Diabet., Jun 2012.
Inter-species differences at the cellular level may also be responsible for the difficulties encountered in modeling irreversible beta cells degeneration in animal models of T1DM.
In the murine beta cell G1/S proteome, the E2F2 transcription factor is abundantly expressed, whereas human islets lack E2F2, but contain E2F3 and E2F7 transcription factors that are absent in murine islets. In humans, E2F3 and E2F7 are involved in regulation of cell cycle progression, proliferation, and metabolic processes, and their dysregulation may contribute to beta cell failure.
The cyclin family members, that regulate the cell cycle progression from G1 to S phase by activating cyclin-dependent kinases, play an essential role in survival and proliferation of beta cells.
The existing inter-species divergences in structure and function of cyclins Fiaschi-Taesch et al., Diab., Jun 2013 are therefore also likely to play a part in the lack of ability of animal models of T1DM to recapitulate the immune-mediated destruction of beta cells.
The stimulus-secretion coupling in human beta cell differs greatly from other species with respect to ion channel composition and function.
The ATP-sensitive potassium (KATP) channel plays an important role in insulin secretion, by linking glucose metabolism-related ATP production to membrane depolarisation. In humans, polymorphisms in the KATP channel subunit genes have been associated with increased risk of T2DM. However, the loss of function in subunits that compose KATP channels does not cause dysregulation of insulin secretion in mice, probably due to unknown species-specific compensatory mechanisms Aguilar-Bryan & Nakazaki, Rec. Prog. Hor. Res., 2011, Rodriguez-Rivera & Barrera-Oviedo, Int. J. Mol. Sci., Apr 2024.
There are also inter-species differences in functional importance of calcium channels that modulate the kinetics of insulin release and glucose-mediated suppression of glucagon secretion Fridlyand et al., Islets, Jan 2013.
The expression of voltage-gated delayed rectifier potassium channels that regulate exocytosis and delayed phase ionic current in humans also differs in rodents, as well as the expression and function of plasma membrane receptor and intracellular ion channel complements facilitating voltage-independent insulin release.
In contrast to mice, human beta cells carry voltage-gated Na+ current via Nav1.6 isoform and Nav1.7, that are important for action potential generation, nociception, and glucose-mediated insulin secretion. This means that Nav1.7 antagonists developed for use as analgesics could have serious implications for human patients who face potential impairment of insulin secretion as a side effect. Such adverse effects of drugs would not be predictable in mice since they lack functional beta cell Nav1.7.
At the tissue level, in humans, skeletal muscle is the primary site of glucose clearance, accounting for 50% to 90% of glucose uptake, making it the primary insulin-sensitive tissue and the primary site of dysregulation in human peripheral insulin resistance. By contrast, liver is the primary site of glucose clearance in rodents, with 5 to 10-fold higher glycogen storage in the liver than in muscle, versus 10-fold more glycogen storage in muscle than in liver in humans. This inter-species difference has functional implications since various aspects of glucose regulation differ between human skeletal muscle and rodent liver Chandrasekera & Pippin, Altex, May 2014.
Glucose homeostasis relies on multiple glucose-sensing cells in the body that monitor blood glucose levels and respond to adjust its glycemia Yoon & Diano, Diabetologia, May 2021.
Hepatocytes express glucokinase that acts as a glucose sensor and can switch between gluconeogenesis and glycolysis based on glucose availability.
Every animal species has a signature blood glucose level or glycemic set point, possibly reflecting evolutionary adaptation. Normoglycemia of one species would be life threatening for another. For example, mouse normoglycemia can be considered diabetic for humans Rodriguez-Diaz, Cell Metab., Mar 2018.
As a consequence of reliance on animal research for insights on glucose regulation, several erroneous concepts were established. For example, in contrast to rodent islets, glucagon input from the alpha cell to the insulin-secreting beta cell is necessary to fine-tune the distinctive human set point.
This and many other inter-species differences have major implications for safety of patients and success of therapies to treat diabetes. For instance, treatments that inhibit glucagon pathways might also eliminate their crucial input to beta cells in humans with diabetes.
Species-specific immunological and genetic differences in T1DM
Species-specific features of innate and adaptive immune systems are also likely to contribute to poor predictive validity of animal models of diabetes.
For example, treatment of T1DM by antithymocyte globulin, that targets multiple T cell antigens, had shown promising results in animal models of T1DM. However, after receiving the same treatment in clinical trials, patients with T1DM had suffered cytokine release syndrome accompanied by a depletion of regulatory T (Treg) cells Gitelman et al., Lancet Diab. & Endocrin., Dec 2013.
The lack of understanding of the precise role of individual components of the immune system in T1DM, combined with existence of species-specific divergencies in the immune system, makes it difficult to know to whether immune-mediated responses in animal models of T1DM recapitulate immune-mediated responses in T1DM patients.
In T1DM, autoreactive CD4+ and CD8+ T cells destroy beta cells, driven by autoantigens like GAD65 enzymes and insulin that are presented by MHC class II on the surface of antigen-presenting cells.
In humans, the proinflammatory cytokines IFN-α, IFN-β, IFN-γ, and IFN-λ, secreted by T cells, macrophage, NK cells and other immune cells, play a major role in mounting of innate and adaptive immune responses to islet antigens Coomans de Brachene et al., Diabet., Feb 2024.
While in humans these cytokines activate the transcription factor STAT4, that plays a pivotal role in insulitis, beta cells apoptosis, and cytotoxicity, the activation of STAT4 is much more restricted in mice Agnello et al., J. Clin. Immun., May 2003, leading to underestimation of its role in T1DM mouse models.
The gold standard non-obese diabetic (NOD) mouse model of T1DM mimics the autoimmune beta cell destruction and STAT4 responses characteristic of human T1DM. Nonetheless, the immune response in NOD mice diverges from that in human T1DM. For example, NOD mice exhibit a stronger Th1 response, leading to direct cytotoxic CD8+ T-cell-mediated beta-cell destruction, whereas in humans, Th17 plays a more significant role, contributing to chronic islet inflammation and beta cell dysfunction, rather than immediate cytotoxicity. The fact that T1DM mouse models may underrepresent Th17-mediated mechanisms is likely to limit their translational relevance.
In addition, insulitis - the inflammation of the islets of Langerhans - shows both qualitative and quantitative species-specific differences between humans with T1DM and animal models of T1DM. In humans, insulitis is less intense and is found in less than 10% of islets. In contrast, the NOD mouse model of T1DM exhibit a more aggressive form of insulitis that touches over 50% of the islets. As a result, reliance on NOD mice may lead to underestimation of the role of Treg cells and environmental modulators that have the potential to slow disease progression in humans.
As part of innate immunity, complement system enhances inflammation, opsonization, and cell lysis. Human studies showed elevated complement components in pancreatic islets of T1DM patients with insulitis. In humans, the complement system is tightly regulated by inhibitors, such as complement factor H (CFH) and factor-H related proteins (CFHR), and their dysregulation could contribute to T1DM Ajjan & Schroeder, Mol. Immun., Oct 2019. However, as genomic studies have revealed, mice lack certain CFHR genes present in humans, such as CFHR-C, Hellwage et al., Immunogen., Oct 2006.
Although the impact of species-specific divergencies in CFH is currently unknown, it could potentially produce inter-species divergencies in complement system regulation, immune response, and severity of insulitis.
The anti-inflammatory cytokine IL-10 suppresses immune responses and promotes Treg function, thereby mitigating beta cell destruction. While in humans, IL-10 is produced during both Th1 and Th2 responses, in mice, IL-10 is much less prominent in Th1 than in Th2 Mestas & Hughes, J. Immunol., Mar 2004.
This inter-species difference in IL-10 production has significant implications for T1DM mouse models, including for the Th1-biased NOD mouse, in which limited Il-10 production leads to a more aggressive disease phenotype than in humans, more rapid onset of T1DM symptoms than in humans, under-represented immune regulatory mechanisms compared to humans and under-estimated human therapeutic responses.
In NOD mice, the binding pocket of the MHC class II allele H2-Ag7 protein, responsible for binding peptides and presenting them on the surface of antigen-presenting cells to T cells, is similar to the binding pocket of the analogous human HLA-DQ. Yet, NOD mice do not express the class II MHC molecule I-E, meaning that they rely solely on I-Ag7 which has a unique peptide binding groove.
The class II human MHC alleles associated with T1DM have a greater degree of polymorphism than the NOD MHC genes, which allows for a more diverse range of antigen presentation Robinson et al., Nucl. Ac. Res., Oct 2019. Unlike in NOD mice, the HLA complex in humans includes several loci that independently confer risk of T1DM Corper et al., Science, Apr 2000.
The efficacy of antigen-specific treatments might differ depending on the MHC alleles involved and the way in which peptides bind in the MHC groove. For example, a single amino acid difference in the B chain of human insulin seems to affect the ability of porcine insulin, but not human insulin, to prevent development of T1DM in NOD mice.
Cluster of differentiation 28 (CD28) is involved in T1DM autoimmune response as a co-stimulatory receptor on T cells that binds to CD on antigen-presenting cells and thereby provides signals for T-cell activation. In T1DM patients, the expression of CD28 infiltrating T cells is elevated.
In mice, CD28 is expressed on all CD4+ and CD8+ T cells, whereas in humans it is expressed on all CD4+ but only about 50% of CD8+ T cells Vuddamalay & van Meerwijk, Front. Immunol., Jul 2017.
Because of species-specific differences in types of CD and surface expression of CD types, the effects on human patients of drugs that target CD are likely to be underestimated or overestimated.
For example, the CD28 antagonist Abatacept showed efficacy in preclinical studies, but not in T1DM patients Orban et al., Lancet, Jul 2011.
In the same manner, the CD28 agonist Theralizumab, also known as TGN1412, caused catastrophic cytokine storm with systemic organ failures in its first human trial, despite being approved as safe in mice and in primates Suntharalingam et al., NEJM, Sep 2006.
Administration of low-dose IL-2, alone or combined with rapamycin, prevented hyperglycemia in NOD mice. Additionally, low-dose IL-2 was reported to cure recent-onset T1DM in NOD mice Grinberg-Bleyer et al., JEM, Aug 2010. However in humans, the same treatment had significant detrimental effects on beta cell function and survival Long et al., Diabetes, Aug 2012.
Natural killer (NK) cells from NOD mice exhibit a defect in functional maturation and have impaired cytotoxic functions, which may render these mice resistant to the toxic effects of IL-2 treatment, leading to failure to predict toxicity to humans.
While a single dose of expanded antigen-specific Tregs could reverse the T1DM phenotype in NOD mice, the attempts to reproduce the same results in T1DM patients have repeatedly failed Bender et al., Sci. Transl. Med., May 2024.
As the above examples show, the reliance on the ability of animal models of T1DM to faithfully recapitulate human insulitis pathology has deleterious repercussions for understanding underlying disease mechanisms and for developing safe and effective therapies for T1DM. Decades of massive failure in clinical trials of hundreds of immune interventions approved in animal models of T1DM have prompted numerous calls for a more human-centred approach Roep & Atkinson, Diabetologia, Oct 2004, Veld, Sem. Immunopath., Jul 2014.
It was suggested that using animal models with a humanized immune system might improve translatability to humans, however, such an approach would face persistent, insurmountable challenges: the role of the human immune system in T1DM is complex and not fully understood, the equivalence of humanized animals to the human immune system was never demonstrated by objective measures Davis, Cell, Dec 2008, and the cross-talk between the human immune system and the rest of human organ systems cannot be recapitulated in animals.
Additionally, another aspect that cannot be recapitulated in humanized animal models, is the development and adaptation of the immune system in response to the environment, including the microbiome, diet and housing. This missing aspect in animal models is crucial, since as twin studies show, the human immune system is more shaped by the environment than by genes Brodin et al., Cell, Jan 2015.
Species-specific differences in tropism for T1DM-associated viruses
Several viruses have been associated with the development of T1DM, particularly through their potential to trigger autoimmune responses that damage insulin-producing beta cells Hober & Sauter, Nature Rev. Endocrinol., Mar 2010, Rothenburg & Brennan, Cell Trends Microbiol., Jan 2020, Altindis et al., PNAS, Feb 2018.
Species-specific genetic variations in host's cell surface receptors and in the immune system have important repercussions for species’ and individuals' responses to viral infections.
For example, humans possess human-specific single nucleotide polymorphisms (SNPs) in IFIH1 gene, that are absent in non-human Ifih1 gene, and that confer human-specific vulnerability (gain-of-function variants) or protection (loss-of-function variants) to T1DM.
The IFIH1 gene encodes the melanoma differentiation-associated protein 5 (MDA5), a cytosolic pattern recognition receptor (PRR), expressed in immune cells and in pancreatic beta cells, that can detect dsRNA from viral infections and trigger an innate immune response.
Human-based genetic and population studies have shown that increased activity of IFIH1/MDA5 in genetically susceptible individuals can promote the development of T1DM, by increasing type I interferon production, beta cells apoptosis, and innate-adaptive immune system crosstalk that subsequently amplifies autoimmune responses Smyth et al., Nature Gen., May 2006, Todd et al., Nature, Jun 2007.
None of the animal species used in T1DM animal research, including NOD/STZ/BB mice and NHP, possess these human-specific SNPs in IFIH1 gene, hindering the study of MDA5-mediated autoimmunity and development of MDA5-targeting therapies Fumagalli et al., PLOS Gen., Nov 2011.
This also raises the question of whether NHP, which do not naturally develop T1DM, possess other protective SNPs in orthologues of human genes involved in T1DM and that could jeopardize safe and effective translation of preclinical studies to T1DM patients.
In humans, coxsackie A virus, coxsackie B virus, echovirus, rubella virus, cytomegalovirus, mumps virus, rotavirus, HIV, SARS CoV2, Hepatitis C virus and others are known to have tropism for pancreatic beta cells and are therefore classified as diabetogenic viruses Rajsfus et al., Cell, Apr 2023.
To experimentally induce symptoms of T1DM in animals, encephalomyocarditis (EMCV), coxsackie, lymphocyte choriomeningitis (LMCV) viruses are typically used. However, there is no evidence that EMCV and LMCV cause diabetes in humans Oberste et al., J. Clin. Microbio., Mar 2000, Jaeckel et al., Ann. NY Acad. Sci., Jan 2006. These viruses are therefore likely to induce T1DM-like symptoms in animal models through mechanisms that are not relevant for human T1DM.
Moreover, in different animal species employed to model T1DM, different viruses may show varying degrees of tropism for pancreatic cells, leading to differences in disease outcomes that hinder translatability to humans Filippi & von Herrath, Diabetes, Nov 2008.
Species-specific differences in gut microbiome
The immune system homeostasis is modulated by the gut microbiome which varies significantly across different species, making findings from microbiological studies in animals not directly translatable to humans.
In diabetes, inflammatory responses can be triggered by an imbalance between beneficial and pathogenic bacteria (dysbiosis), by an increased intestinal permeability to bacterial endotoxins, by bacterial stimulation of pro-inflammatory cytokines, or by elevated levels in the blood of gram-negative bacteria cell walls-derived lipopolysaccharides.
In humans with T1DM, it has been reported that the proinflammatory environment in the gut is associated with the combination of an increased relative abundance of Bacteroidetes and a decreased level of Firmicutes, regardless of geographical location Jamshidi et al., Gut Path., Oct 2019.
Studies from human cohorts suggest that dysbiosis contributes to T1DM primarily by promoting Th17/Th1 autoimmunity, gut barrier permeability, and beta cells destruction Kostic et al., Cell Ho. & Micr., Feb 2015, Vatanen et al., Nature, Oct 2018.
Dysbiosis of the gut microbiota is suggested to occur early in life, aggravating gut inflammation and influencing the immune system, before the onset of T1DM. The relationship between T1DM autoimmunity and gut microbiome dysbiosis is likely to be bidirectional and needs to be further investigated using human-based metagenomics and metaproteomics Durazzo et al., J. Clin. Med., Nov 2019.
The study of the microbiome has historically relied on mouse studies, based on coarse comparisons at higher taxonomy hierarchy level with a poorly characterized mouse gastrointestinal (GI) microbiota. However, advances in metagenomics, that have enabled a much more granular view of mouse microbiota at lower taxonomy level, have shed light on previously unknown profound species-specific differences in GI microbiome composition, quantity, and function.
A comparative survey of the phylogenetic composition of 16 human subjects and 3 often used mouse lines indicated that their microbiota show considerable similarity at the genus level but is quantitatively very different Hugenholtz & de Vos, Cell. Mol. Life Sci., Nov 2017.
Comparison of comprehensive mouse microbiota genome to microbiota from the human GI genomes shows an overlap of 62% at the genus but only 10% at the species level, demonstrating that human and mouse gut microbiota are largely distinct Kieser et al., Plos, Mar 2022.
What is more, mouse and human microbial strains of the same species can have divergent gene content and function, as is exemplified by the species Limosilactobacillus reuteri which has mouse-adapted and human-adapted strains, however, with very different functions.
Previously, DNA sequencing of fecal samples of mouse strains of diverse genetic, provider, housing and diet backgrounds and comparison to the human gastrointestinal genome has demonstrated that only 4% of mouse GI microbial genes were shared with human GI microbial genes Xiao et al., Nature Biotech., Sep 2015.
Rats tend to have a higher abundance of the phylum Bacteroidetes compared to mice. Mice, on the other hand, have a higher abundance of the family Muribaculaceae within the phylum Bacteroidetes Nagpal et al., Front. Microbiol., Nov 2018.
Even within closely related species, like striped hamsters and Djungarian hamsters, there can be differences in bacterial families and genera that reflect their distinct lifestyles and dietary habits Fan et al., Curr. Microbio., Mar 2023, De Jonge et al., Front Microbiol., Jun 2022.
Although the gut microbiota in humans was closer to NHP than to mice and rats, there are still significant differences between the two species.
For example, African green monkeys show an opposite microbial taxa and genes responses to a typical high-protein, high-fat, low-fiber Western diet compared to humans, suggesting that NHP are not a good model for studying the effect of the human gut microbiota on host metabolism and microbiome-associated human diseases Amato et al., Microbiome, Nov 2015.
Efforts have been made to humanize mice with human microbiota and to inter-cross inbred strains. However, it was found that the GI microbiome in mice varied immensely depending on the housing conditions, even more so than on the genetic background, and that the inter-individual variation in GI microbiome decreased after 10 generations Hugenholtz & de Vos, Cell. Mol. Life Sci., Nov 2017.
The so called human microbiota-associated (HMA) mouse models, that are designed to study the contribution of a dysbiotic microbiome to development of human diseases by comparing disease phenotypes of germ-free mice colonized with the fecal microbiota of patients to those of mice colonized with the microbiota of healthy controls, do not replicate patients' microbiome Arrieta et al., Cell Host Micr., May 2016 Engrafted HMA mice showed only a partial resemblance to the donor microbiota. It is believed that only the microbiome phylotypes that are adapted to the mouse intestinal environment, and that survive an ecological shift after engraftment to mice, will remain in the mouse gut.
Human-based research has shown that the gut microbiome plays a crucial role in shaping both the innate and adaptive immune system from infancy through adulthood through human host-specific interactions between human host-specific immune system and human host-specific microbiota.
Therefore it is not surprising that HMA mice have a low immune maturation with reduced numbers of adaptive and innate intestinal immune cells when compared with mice that harbor a murine microbiota.
That means that the engrafted human microbiota is not representative of the microbiome compositions in patients', neither does it reflect human pathologies-associated dysbiosis and immune responses.
Face validity - How well do animal models replicate the human disease phenotype?
Since the end of the 19th century, several dozen animal species and strains were used to model human Type 1 diabetes mellitus (T1DM) with the goal to probe disease causes and risk factors, study disease mechanisms, develop new therapies and assess safety and efficacy of therapeutic candidates.
None of these T1DM animal models have faithfully and fully recapitulated the pathophysiology of T1DM, nor the inter-individual heterogeneity of disease phenotypes Singh, Front. Endocrinol., Feb 2024, Pandey et al., Biomed., Oct 2023.
Methods so far employed to induce symptoms of T1DM in animals include selective breeding of strains with spontaneous symptoms, gene editing, chemical induction, viral induction, and surgical induction.
With time and growing knowledge of advantages and drawback of each animal model, the animal research community had developed specific animal models of T1DM tailored to each induction type with the aim to recapitulate specific subsets of diabetes pathology, identify mechanisms underlying specific symptoms and select potential drug targets.
However, there are several major limitations to this approach and that cannot be addressed whatever the animal species, experimental manipulation and diabetes induction method used.
The most important being that fundamental differences between humans and animals in pancreas anatomy, physiology, genetic background, gene expression, immune system, diet, viral tropism and environmental exposure, that work together in a complex interactive manner, represent a major barrier to extrapolation of findings to humans.
A narrow focus on specific pathological pathways and specific features of diabetes in specific species/strains to address specific questions is another critical limitation of animal models, aggravated by the fact that certain animal models recapitulate some T1DM symptoms in some experimental conditions and not in others, making it impossible to gain understanding of other diabetes-related pathways, to compare findings from different species/strains and to learn how individual pathways fit into the whole picture.
Streptozotocine (STZ), alloxan and cyclophosphamide are among the most commonly used chemicals to induce T1DM.
Although pigs and NHP present with a pathophysiology that is more similar to humans with T1DM, they are particularly complex and costly to maintain in comparison to rodents.
At low doses, STZ can induce an immunological and inflammatory response, while a high dose STZ results in significant pancreatic beta cells death and insulin deficiency.
In this method, hyperglycemia develops mainly through direct cytotoxic action on beta cells rather than through a number of intermediate steps leading to autoimmunity.
Also, toxicity of chemicals may not be limited to the pancreas and can affect other organs, creating confounding adverse effects, higher morbidity and high mortality in animals.
Another drawback of this method is difficulty in producing irreversible T1DM in rabbits, dogs, pigs and NHP. Moreover, dogs may develop only moderate hyperglycemia without change in body weight or in blood insulin levels.
The gold standard non obese diabetic (NOD) mouse is a polygenic model susceptible to autoimmune diabetes, developed by selective breeding of mice that develop spontaneous diabetes. This mouse model displays hyperglycemia, insulitis, destruction of beta cells as well as several diabetic complications, such as neuropathy, nephropathy and retinopathy Aldrich et al., Cell Transpl., Aug 2020. Of note, the frequency of T1DM in NOD females is 3-fold higher than in NOD males, whereas the prevalence of T1DM in the human population is slightly higher in males than in females.
The commonly used Akita mouse is a spontaneous model that carries a heterozygous missense mutation in the insulin 2 gene that ultimately results in endoplasmic reticulum stress, pancreatic beta cell destruction, insulin deficiency, and hyperglycemia.
Unlike in human T1DM, the Akita mouse pathophysiology is not related to an autoimmune process and does not show insulitis, which limits its use for studying autoimmune aspects of T1DM. Moreover, Akita mice have a greater capacity for beta cell regeneration compared to humans, which is likely to lead to overestimation of the potential of drugs to halt and reverse beta cell degeneration.
While the BB rat displays spontaneous autoimmune destruction of beta cells, hyperglycemia and ketoacidosis, it is also associated with T-lymphopenia, which is not a hallmark of T1DM in humans.
The LETL rat manifests spontaneous autoimmune destruction of beta cells without T-lymphopenia, however at a low incidence of 20% which makes it an unreliable model for T1DM.
Infection with Encephalomyocarditis, Coxsackie and Lymphocyte choriomeningitis viruses induces immune mediated-destruction of beta cells in mouse, rat, hamster and NHP models of T1DM. However, virally-induced T1DM animal models have systemic effects that are not typical of T1DM in humans, such as pneumonia, gastrointestinal disorders, and myocarditis, and it is unclear whether findings in virally-induced T1DM animal models can be applied to humans with T1DM.
Surgical procedures, such as pancreatectomy, islet transplantation and thymectomy, employed to induce T1DM in mice, rats, rabbits, dogs, pigs and NHP, mostly elicit moderate hyperglycemia followed by pancreatic regeneration, which does not represent a realistic scenario for humans with T1DM.
Removal of thymus to induce autoimmune responses in the pancreas can lead to broader immune deficiencies which are likely to confound results of studies in thymectomy-derived animal models of T1DM.
Moreover, these surgical procedures can induce surgical injury, stress and inflammation which do not reflect the human T1DM pathophysiology.
The inability of animal models of T1DM to fully and faithfully recapitulate T1DM phenotype and pathophysiology can be explained by numerous species-specific differences in structure and in function of pancreatic islets, in glucose regulation, in gene expression regulation, in immune systems, in tropism for T1DM-associated viruses and in gut microbiome.
Construct validity - How well do the mechanisms of disease induction in animals reflect the currently understood etiology of the human disease?
Surgical induction as method to model T1DM/T2DM in animals clearly does not correspond to any of the causes of T1DM and T2DM observed in patient populations.
Pancreatectomy does not allow to model the insulin resistance in T2DM. It can mimic to some extent the loss of pancreatic beta cells, but not through autoimmune mechanisms characteristic of T1DM.
Insulitis and inflammatory response to surgical injury are very different in causes and processes. Artificial induction of T1DM through surgery is therefore not an appropriate method for studying the mechanisms that underly insulitis in T1DM.
Thymectomy can lead to impairment of adaptive immune processes to islet antigens, which represents a major barrier for recapitulating the human-relevant mechanisms that underpin T-cell-mediated damage to pancreatic islets. To date, there has been only anecdotal clinical evidence of thymectomy resulting in increase of autoimmunity Al-Bkoor et al., JCEM Case Rep., Mar 2024. Given that thymectomy is not considered to be a direct cause of diabetes in the general patient population, thymectomy-derived animal models cannot be relied upon to gain human-relevant knowledge on mechanisms of T1DM.
Renal denervation can reduce sympathetic nervous system activity and interfere with mechanisms of T2DM by improving insulin sensitivity and glycemic control in animal models. Moreover, these effects are at odds with evidence from clinical trials Koutra et al., Acta Diabet., Dec 2023.
Another drawback of surgical induction methods is the risk of accidental removal of exocrine acinar cells during surgery, which can lead to malabsorption and nutritional deficiencies Capurso et al., Clin. Exp. Gastroenterol., Mar 2019, indirectly producing confounding effects on glucose metabolism and insulin sensitivity.
In chemically-induced animal models, beta cells are degraded through direct cytotoxic action of STZ and alloxan. However, since it is widely believed that T-cell mediated autoimmunity occurs prior to beta cells degradation, this method is not consistent with the current state of knowledge. What is more, induction of diabetes with chemicals can trigger off-target oxidative stress, cellular and tissue toxicity, making it difficult to distinguish between mechanisms that are relevant for diabetes from those that are not.
Dozens of human genetic polymorphisms that contribute to the risk for T1DM/T2DM cannot be recapitulated in animal models of T1DM/T2DM. Such experiments would be very long and would still exclude the effect of human-specific genetic background.
Moreover, genetically altered animal models cannot recapitulate the complex interplay of genetic, environmental and lifestyle factors.
Despite many similarities in disease phenotype that make the NOD mouse the animal model of choice for T1DM, the NOD mouse presents with major species-specific differences, including in the genetic background, immune system, diet, microbiome, and susceptibility to T1DM-associated viruses. As a result, the mechanisms of T1DM in NOD mice are likely to differ from mechanisms of T1DM in humans.
Crucially, the NOD mouse is not predictive of treatment efficacy in T1DM patients. For instance, several immunotherapies designed to treat the autoimmune-mediated pathogenesis in T1DM had shown promising results in NOD mice, only to be discarded in clinical trials.
Predictive validity - How well do animal models predict safety and efficacy of therapies in patients?
The predictive validity of animal experiments for endocrine diseases is poor and below the 7.9% average for all indications BIO, New Clin. Dev. Succ. Rat., 2011-2020.
The likelihood for clinical approval of drugs for endocrine diseases was only 6.6% over 2011-2020.
While the discovery of the insulin therapy through experiments in dogs marked a landmark breakthrough more than a century ago, it stands as a rare exception. In the decades since, the sacrifice of millions of animals to discover new treatments for diabetes has not led to medical advances of added value for T1DM patients.
Insulin administration through daily injections, or an insulin pump, remains to date the mainstay of treatment Sapra & Bhandari, StatPearls, Jun 2023.
Despite glucose control with insulin therapy, many T1DM patients develop cardiovascular disease, neuropathy, retinopathy, chronic kidney disease and
The therapy itself aims to compensate for the missing insulin but does not halt the autoimmune-mediated destruction of pancreatic beta cells.
Numerous pharmaceutical agents prevented and even reversed T1DM in NOD mice, yet, these successes were not replicated in clinical trials. Although some interventions allowed to delay the onset or progression of disease in certain subsets of patients, none have resulted in a cure.
After over 20 years in the clinic, Provention Bio’s CD3 antibody teplizumab was approved by the FDA in 2022 as treatment for delaying the onset of T1DM Mullard, Nature Rev. Drug Disc., Dec 2022.
The overwhelming majority of therapies that had shown promising results in T1DM animal models, including GAD65(alum) antigen-specific immunotherapy, dipeptidyl peptidase-4 inhibitor sitagliptin combined with proton pump inhibitor lansoprazole, and IL-1 inhibitors/IL-1 receptor antagonists, failed to demonstrate meaningful benefits in clinical trials.
In some cases, therapies approved in preclinical trials have even worsened patients' condition. One such example is the rapamycin/interleukin-2 combination treatment. Effective in treating T1DM in NOD mice, administration of rapamycin/interleukin-2 in clinical trials resulted in deterioration of beta cells function in patients Long et al., Diabetes, Aug 2012.
Another major problem of animal research is that therapies that have the potential to save patients' lives may not show benefit in animal models. One such example is abatecept that had significantly improved C-peptide response in patients with new-onset T1DM but had shown completely opposite results in NOD female mice Reed & Herold, Nature Rev. Endocrinol., Jan 2015.
In conclusion, prevention and treatment of T1DM remain suboptimal, with large inter-individual variations in responses to treatments. In clinical trials for antithymocyte globulin, for instance, half of participants with stage 2 T1DM remained diabetes-free for up to four years, while the other half progressed to stage 3 within two months Foster et al., Diab. Care, Dec 2023.
Ethical validity - How well do animal experiments align with human ethical principles?
Preclinical
Ethics is a human-specific philosophical concept. Animal experimentation is unethical in essence by human standards, since it involves physical constraint, psychological suffering and deprivation of freedom, social interactions, natural environment, and life purpose.
In addition to this baseline, animal experiments inflict severe clinical harm in animals De Vleeschauwer et al., Animals, Aug 2023:
Table S5: Severity classification of chemical disease models
Diabetes: causing up to severe clinical signs
Table S13: Severity classification of genetically altered (GA) lines
GA lines with diabetes like NOD mice, BB rats: Severe
GA lines resulting in long-term moderate pain or short-term severe pain: Severe
GA lines resulting in long-term moderate anxiety or short-term severe anxiety: Severe
Table S4: Severity classification of clinical signs
Skin wounds - Causing up to severe clinical signs: Mice: >10 mm body, >3 mm face - Rats: >30 mm body, >10 mm face; Skin open with signs of infection (wet discharge of blood or pus) or open to muscle or bone.
Table S3: Severity classification of surgery and surgical induction of disease
Major surgery causing permanent or progressive loss of function of specific organs/senses/body systems; Organ/cell transplantation or device testing where rejection/failure may lead to severe distress, death or impairment of the general condition of the animal: Severe
Surgical complications resulting in lethality
Table S6: Severity classification of infectious diseases
Viral diseases : up to severe clinical signs or long-lasting moderate clinical signs
Clinical
While there is no consensus on whether an unethical act can be justified by a pursuit of a hypothetically ethical outcome, it was suggested that animal research was necessary to advance safe and effective treatments for human diseases.
However, statistics consistently show that clinical success rates of drugs developed and tested in animals is very low, raising the question of whether it is ethical to put the health of patients at risk.
Over 2011-2020, the Phase I to II transition success rate, that primarily focuses on assessing the safety and tolerability of drugs, was 43.3% for endocrine diseases, well below the 52% average of all indications BIO, New Clin. Dev. Succ. Rat., 2011-2020.
Phase II to III transition success rate for endocrine diseases group was 26.6% versus 28.9% average for all indications, highlighting the low effectiveness of drugs candidates developed in animal models of human endocrine diseases.
Intrinsic validity - How well do animal models capture the clinical heterogeneity of the human disease?
The heterogeneous pathogenesis and phenotypes of Type 1 diabetes mellitus (T1DM) are not recapitulated in animal models of T1DM.
Extrinsic validity - How well does animal experimentation generate reliable and reproducible outcomes?
It is often argued that although animal models have severe limitations, experimenting on animals enables to gather insights that may be valuable. However, the basic precondition for a hypothetical benefit is not met since the majority of animal experiments is irreproducible Freedman et al., PLOS Biol., Jun 2015.
Contributing factors include flawed experimental design, variation in animal strains and experimental conditions, and lack of transparency on methodology and results of animal studies.
In a bid to improve the quality of reporting of animal experiments, the ARRIVE - Animal Research: Reporting of In Vivo Experiments - guidelines, were published in 2010 and updated to ARRIVE 2.0 in 2020 Arrive guidelines website.
Nonetheless, and in spite of significant investment in dissemination, various incentives and training of animal researchers, the Arrive guidelines remain poorly implemented Percie du Sert et al., BMC Vet. Res., Jul 2020, Bazoit, BioRxiv, Feb 2025.
As a result of weak relevance, rigor and reliability of animal studies, erroneous and misleading hypothesis are generated, animal and human lives are needlessly sacrificed and dozens of billions of dollars are annually wasted Yarborough et al., PLOS Biol., Jun 2018.
Beyond the problem of experimental design and reporting of animal studies, there are deep-seeded cultural reasons that are not likely to be addressed any time soon, such as the "publish or perish" culture that discourages from unbiased analysis and reporting of negative results and rewards exaggerated and overhyped claims Smaldino & McElreath, Roy. Soc. Op. Sci., Sep 2016.
In Summary
Type 1 Diabetes Mellitus (T1DM) is an autoimmune disease characterized by chronic hyperglycemia. It is associated with an increased risk of cardiovascular disease, nephropathy, retinopathy, diabetic wounds, psychiatric disorders, dental issues and peripheral neuropathy.
Within individuals with T1DM, epidemiological, clinical, genetic, immunological, histological and metabolic differences suggest heterogeneity in pathophysiological mechanisms.
In vivo modeling of T1DM causes severe suffering in animals. Yet, owing to human-specific features of pancreatic anatomy and function, glucose regulation, genetic background, gene expression regulation, and immune system, animal models of T1DM do not faithfully recapitulate T1DM phenotypes, pathophysiology, and response to treatments.
To date, there is no cure for T1DM and treated patients often go on to develop cardiovascular, renal, and skin ulcer complications with potentially severe consequences.
A human-centric approach to research is needed to advance understanding of disease mechanisms and discovery of innovative treatments to prevent, treat, and reverse heterogenous T1DM endotypes.
How is Human-Based In Vitro the Answer to Advance Biomedical Research into Type 1 Diabetes Mellitus
*To model heterogenous T1DM pathophysiology and phenotypes, using patient-derived 3D tissues/pancreas organoids/pancreas-on-chip.
*To determine the role of genetic polymorphisms in T1DM, by gene editing (overexpression, knock-out, knock-down) and gene perturbation (CRISP interference, siRNA) in human 3D pancreas tissues/organoids/pancreas-on-chip.
*To identify epigenetic drivers of pancreatic beta cells destruction and to test potential epigenetic therapies for T1DM, using epigenetic editing (CRISPR activation/repression, lncRNA) in T1DM patient-derived pancreatic 3D tissues/organoids/multi-organs-on-chip.
*To determine the contribution of individual and combined inducers to T1DM, by supplementing the culture medium with nutrients and chemicals in human healthy 3D pancreatic tissues/organoids/(multi)organs-on-chip Zbinden et al., Matrix Bio., Jan 2020. To determine individuals’ susceptibility to a certain inducer type, using T1DM patient-derived pancreatic 3D tissues organoids/(multi)organs-on-chip with patient-specific genetic background.
*To model human pancreatic beta cells-targeted autoimmunity, and study the crosstalk between innate and adaptive immunity in T1DM, using immunocompetent T1DM patient-derived iPSCs/pancreatic 3D tissues/organoids/pancreas-on-chip Leite et al., Cell Rep., Jul 2020, Armitage et al., Front. Endocrinol., Nov 2021, Joshi et al., Diabetol., Sep 2019.
*To study how different dietary components influence the gut microbiome, using human/patient-derived in vitro intestine and ex vivo fermentation systems Cieplak et al., FEMS Microbiol. Lett., Sep 2018, O'Donnell et al., Front. Microbiol., Aug 2018, Pantoja-Feliciano et al., BMC Microbiol., Jan 2023.
*To determine the role of the gut microbiome in T1DM pathogenesis and response to therapies : microbiome-host interactions, role of individual gut microbes, microbiome dysbiosis etc., by using immunocompetent T1DM patient-derived tissues/pancreas-gut-microbiome-on-chip.
*To study the metabolic and endocrine mechanisms of glucose regulation in T1DM, by recapitulating the human-relevant crosstalk between the pancreas and other organs, such as liver, gut, adipose tissue, and skeletal muscle, in patient-derived iPSC multiorgan-chip Jeon et al., Biomed. Microdev., Sep 2020.
*To determine the capacity of T1DM-associated viruses to trigger T-cell mediated autoimmunity, by infecting immunocompetent human pancreatic tissues/organoids/pancreas-on-chip with human diabetogenic viruses Yang et al., Cell St. C., Sep 2024, Dechaumes et al., Microorg., Sep 2020.
*To uncover mechanisms of human T1DM-related wound healing impairment and to develop new treatments for diabetic ulcers, using immunocompetent T1DM patient-derived 3D skin models/skin organoids/skin-on-chip Smith et al., Tiss. Eng., Feb 2021, Ejiugwo et al., Tiss. Eng., Feb 2021.
*To study pathogenic mechanisms that are distinct and common to each T1DM endotype, using genomics, epigenomics, transcriptomics, microbiomics and proteomics AI-powered analysis in large scale patient-derived pancreatic 3D tissues /organoids/pancreas-on-chip, and by linking multi-omics data to clinical and biochemical patient data. To discover new therapeutic leads that target pathogenic pathways.
*To study mechanisms of T1DM diabetic nephropathy, retinopathy and peripheral neuropathy in healthy/patient-derived human kidney, retinal and neuronal organoids/(multi) organs-on-chip Achberger et al., Elife, Aug 2019.
*To find new predictive biomarkers for real-time assessment of response to treatments, using organs-on-chip with integrated sensors Clarke et al., Sensors, Feb 2021.
*To test efficacy of drug candidates for T1DM by high-throughput screening in patient-derived 3D tissues/organoids/organs-on-chip.
*To test safety and efficacy of single and combination therapies for T1DM in patient-specific organoids/organs-on-chip, in a personalized medicine approach.
Although in vitro methods have inherent limitations, their relevance to human biology far exceeds that of animal research.
Animal model organisms were never comprehensively compared to humans and scientifically validated. Complementing in vitro methods with animal experiments is not effective for human patients, because species-specific differences prevent reliable integration and translation of results to humans.
While animal research benefits from experimenting on a complete organism, model organisms fail to replicate the interplay of thousands of human-specific features, from molecular level to organism level, and are therefore not representative of the complete human organism.
To address the challenges of individual human-based in vitro models, they can be integrated with other human-based in vitro methods, AI-driven analysis, clinical data, and real-world patient data.
Have you leveraged in vitro methods in unique ways? We would love to hear how! Join the conversation to exchange ideas, collaborate and inspire new directions in human-based science!