Skip to main content
Intended for healthcare professionals
Free access
Research article
First published online September 22, 2016

Regulatory Forum Opinion Piece: Review—Toxicological Pathology Profile and Regulatory Expectations for Nonclinical Development of Insulins and Insulin Analogues

Abstract

The toxicological profile of insulins is exclusively due to exaggerated pharmacology resulting in hypoglycemic findings. Insulin analogues displaying modifications and aimed at improving pharmacokinetics do not induce different toxicity. The main target is the brain displaying neuronal necrosis. Wallerian degeneration of nerves occurs rarely after severe hypoglycemia. These findings are of potential human relevance; nevertheless, these changes are induced in normoglycemic animals whereas diabetic patients suffer from hyperglycemia. Therefore, it is usually not difficult to achieve a therapeutic window for subsequent use in patients. Based upon this and in the absence of classical toxicity, there has been no scientific need for diabetic animal models. A greater challenge is the mitogenicity already inherent with regular insulin. Thus, the focus for preclinical safety evaluation of analogues is to demonstrate that modifications in regular insulin do not result in enhanced mitogenicity. The approaches used to assess the mitogenic potential of insulin analogues have changed over time driven by scientific progression and changes within the regulatory environment. Therefore, in vitro and in vivo evaluation of cell proliferation has become common practice, and to date there has been no evidence that the mitogenic potential of insulin analogues may be increased compared to regular insulin.
Diabetes mellitus, along with its vascular complications, is still one of the primary causes of increased cardiovascular morbidity and mortality. Since their first successful use, about 100 years ago, insulins have been increasingly and widely administered for the treatment of both type 1 and 2 diabetes in order to achieve normal blood glucose levels.
Based on the substantial progress in molecular biology and gene technology, biosynthetic human insulin homologues and analogues have now become available. This developmental trend starting in the 1980s represented a revolutionary step in the treatment of diabetes. Insulin analogues displaying modifications in amino acid sequence and in peptide structure enabled the optimization of the pharmacokinetic profile of exogenous insulin to the physiological pattern of glucose release in patients.
All of these advancements in the successful treatment of diabetes in man were made possible based upon a comprehensive preclinical evaluation of both efficacy and safety. Contrary to the preclinical pharmacological evaluation of insulins/insulin analogues that have been widely published, to our knowledge, the toxicological profile and issues related to the preclinical safety assessment of insulins/insulin analogues have not been reviewed so far in the literature.

Structure and Pharmacology of Insulin

Insulin is a polypeptide composed of 51 amino acids. They are arranged on 2 chains (A and B) comprising 2 interchain disulfide bonds and 1 intrachain bond on chain A (see also Figure 1). This pancreatic hormone originating from the Islets of Langerhans is crucial for the regulation of carbohydrate and fat metabolism as indicated below.
Figure 1. Sequence and structure of human regular insulin. Amino acid sequence modifications of engineered insulin analogues: long-acting insulin analogues: insulin glargine—asparagine is replaced by glycine at A21 and 2 asparagines added to the C-terminus = B31 and 32, insulin detemir—threonine at B30 is removed and a myristic acid is linked to lysine at B29, insulin degludec—threonine at B30 is removed and a hexadecanedioic acid is linked to lysine at B29. Fast-acting insulin analogues: insulin lispro—proline and lysine are reversed at B28 and B29, insulin aspart—proline is replaced by aspart at B28, insulin glulisine—asparagine is replaced by lysine at B3 and lysine by glutamine at B29.
The endocrine pancreatic hormone released from the β cells of islets of Langerhans is crucial for the regulation of carbohydrate metabolism as indicated in Figure 2. In addition to glycemic control, there are effects of insulin on fat metabolism, and these include increased lipogenesis and increased uptake of lipids from blood. This is crucial for treatment of type 2 diabetes, because high levels of low-density lipoprotein cholesterol, triglycerides, and fatty acids contribute significantly to insulin resistance and metabolic syndrome.
Figure 2. Pharmacological effects of insulin: insulin binds to its receptor (1). Complex phosphorylation cascades leading to translocation of glucose transporter type 4 to the plasma membrane (2). Glucose uptake into liver, skeletal muscle, and adipose tissue cells (3). Insulin stimulates glucose “consumption” by hepatic glycogen synthesis (4), glycolysis (5), and suppression of hepatic gluconeogenesis as well as stimulation on triglyceride synthesis (6).

Insulin Analogues

The disadvantages of conventional insulin therapy in the past included antigenicity and impurity of insulins from animal sources and the fact that exogenous insulin delivery did not reflect the daily dynamic of blood glucose levels related to food intake, thus conventional insulin therapy did not manage postprandial blood glucose peaks in an optimal way (Figure 3). Based on the substantial progress in molecular biology and gene technology, biosynthetic human insulin homologues and analogues became available. This trend in development starting in the 1980s represented a revolutionary step in the treatment of diabetes. Insulin analogues displaying modifications in amino acid sequence and in peptide structure allowed the optimization of the pharmacokinetic profile of exogenous insulin to more closely follow the physiological pattern of glucose release in patients (see reviews from Owens, Zinman, and Bolli 2001; Owens 2002; Hirsch 2005; Evans et al. 2011).
Figure 3. Endogenous insulin secretion closely follows the circadian dynamic of blood glucose which is influenced by food intake. Postprandial blood glucose peaks are managed by synchronous insulin release.
Recombinant deoxyribonucleic acid (DNA) technology revolutionized insulin development and allowed the removal of certain former disadvantages by producing biosynthetic human homologues with less antigenicity and higher purity and genetically engineered insulin analogues with modified amino acid sequence. The latter allowed treatment to be closer to circadian kinetics and dynamics of endogenous insulin secretion. Furthermore, amino acids were exchanged, deleted, or added; and fatty acids were attached (insulin detemir and degludec) changing the structure, charge, solubility, and self-association propensities which altered the pharmacokinetic/dynamic profile (Figures 1 and 4).
Figure 4. Pharmacokinetic profile of fast-acting insulin analogues (lispro, aspart, glulisine) and long-acting insulin analogues (glargine, detemir, degludec) compared to conventional insulins.
Long-acting insulins have modifications in amino acid sequence that result in increased hexamer formation, decreased release into systemic circulation, and prolonged pharmacodynamics which maintain baseline levels without peaks and drops and results in overall glycemic control throughout the day.
Fast-acting insulins (rapid and short acting) have modifications in amino acid sequence that result in decreased hexamer formation, increased release into systemic circulation, and a rapid onset and short duration of activity. They display a time activity profile with a short activity peak that matches physiological mealtime requirements and cuts off postprandial hyperglycemia.
The use of combinations of long- and fast-acting insulins provides a pharmacokinetic and pharmocodynamic profile with improved simulation of the circadian kinetic and dynamic profile of endogenous insulin in relation to and between mealtime glucose levels. This in turn provides improved glycemic control and decreases hypoglycemic risk.
Tailoring the pharmacokinetic and pharmacodynamic profiles of insulin analogues to the physiological pattern of glucose release in patients has allowed for more precise treatment of diabetic patients. In this review, we discuss the toxicological profile and issues related to the preclinical safety assessment of insulins/insulin analogues.

Material and Method

Test Animals

Rat and dog studies performed at Sanofi and cited in this review article were authorized by the competent authorities according to the German Animal Welfare Acts and conducted in compliance with the European Economic Community (EEC) Council Directive of the European Community from November 24, 1986, on the approximation of laws, regulations, and administrative provisions of the member states regarding the protection of animals used for experimental and other scientific purposes (86/609/EEC).
Five- to nine-week-old Wistar (Hoe: WISKf) or Sprague-Dawley rats (Charles River Laboratories, Harlan Winkelmann GmbH, Borchen, Germany) were used; they were separated by sex, housed with a maximum of 3 animals per cage, and subjected to a 12-hr light–dark cycle in air conditioned rooms. Unlimited access to food (Altromin® 1324, Lage, Germany; ssniff® R/M-H pellets, Soest, Germany) and water was provided.
Eight- to 14-month-old beagle dogs of different strains (HsbCpd: DOBE, HsdBor. BEAG, Marshall, Hoe: BEAK) were housed in animal facilities with temperature at 18°C. Each dog was kept in a separate kennel with access to an outdoor exercise area. Different standard diets were utilized (ssniff Hd-H, Pedigree® for puppies, Pedigree PAL, Soest, Germany) before treatment, and tap water was provided ad libitum.

Test Article and Toxicity Studies

The design of 7 Sanofi studies with insulins and insulin analogues cited in this review article are described in detail in Table 1 together with glucose levels, clinical observations, and target organs.
Table 1. Target Organs in Relation to Dose Levels, Clinical Observations, and Glucose Levels.
Study type Dose levels in IU/kg/day Target organs at high dose Clinical observations at high dose Glucose levels in mmol/L at high dose Glucose levels in mg/dl at high dose (conversion)
2-Week study in rats 50–100–200 Nerves Limited use of hind limbs; premature euthanasia/mortality 0.67–3.46 12.1–62.3
1-Month study in rats 2–10–50 Brain and nerves Limited use or loss of use of hind limbs, uncoordinated gait, premature euthanasia/mortality 0.89–4.27 16.0–76.9
1-Month study in rats 20–50–100 Brain Not specified (poor clinical conditions), premature euthanasia/mortality 1.60–3.12 28.8–56.2
1-Month study in dogs 0.5–1.0–1.5 Brain and nerves Trembling; twitching, mobility disorders, premature euthanasia 2.5–4.7 45.0–84.6
6-Month study in dogs 0.5–1.0–2.0 Brain Convulsions, premature euthanasia 1.7–2.9 30.6–52.2
12-Month study in rats 5–10–40–100 (2.5–5–20–50 twice daily) Brain Hypoactivity, uncoordinated gait, convulsions, premature euthanasia/mortality 1.3–2.2 23.4–39.6
2-Year study in rats 2–5–12.5 Brain Not specified (poor clinical conditions), premature euthanasia/mortality Not determined Not applicable
Test articles were administered subcutaneously (sc) to rats or dogs once or twice daily for study durations of 2 weeks to 2 years, and sc to dogs once daily for study durations of 2 weeks to 6 months. The sc route was chosen because it is the intended human therapeutic route of administration. All studies with durations of ≥1 month were performed in accordance with the principles of Good Laboratory Practice. In some of the 2 week rat and dog studies, a recovery period of 2 weeks without treatment was present in order to assess the reversibility of potential treatment-related changes. Studies referenced in this article were performed from 1993 to 2010.

Toxicological Profile of Insulin and Insulin Analogues

Owing to the peptide structure of insulins, it was not surprising that classical chemical toxicity is absent. The toxicological profile is not only dominated by exaggerated pharmacodynamics at suprapharmacological dose levels resulting in hypoglycemia (blood plasma glucose ≤ 3.9 mmol/L; American Diabetes Association [ADA] 2005), it is exclusively due to the effects of hypoglycemia.
As for regular insulins, the toxicological profile of insulin analogues is also dominated by findings caused by exaggerated pharmacologic activity resulting in hypoglycemia.
According to the literature, all insulin analogues tested so far have not displayed a toxicological profile different from that of regular insulins.

Target Organs of Insulins and Insulin Analogues in Preclinical Toxicity Studies

Overview on Repeated Dose Toxicity Studies Evaluated in This Review

Central nervous system (CNS) findings related to suprapharmacologic insulin levels

In our own repeated dose toxicity studies in nondiabetic, normoglycemic rats, the brain was the main target organ of hypoglycemia and displayed prominent, distinct, and reproducible neuronal findings. Independent of the use of regular insulin or an insulin analogue, the hippocampus was particularly affected. Based on the subtopography (Figure 5), Cornu ammonis 1 (CA-1) to CA-3, neurons of the CA-1, and the crest of dentate gyrus have been selectively affected showing neuronal necrosis. In the hippocampus, there was a severity gradient starting from the subiculum and medial CA-1 becoming more attenuated toward the lateral CA-1. A sharp demarcation between damaged and normal areas was usually present. Affected neurons displayed eosinophilic and shrunken nuclei and cytoplasm. Neuronal necrosis was frequently accompanied by vacuolation of the neuropil (Figures 6A and B and 7A and B). Accompanying inflammatory responses including glial reaction were not observed, and lesions were not necessarily bilaterally symmetric.
Figure 5. Topography of the hippocampus area: CA = cornu ammonis; DG = dentate gyrus; sub = subiculum. H&E, original objectives 6.3×.
Figure 6. Hippocampus of a rat treated with an insulin analogue at 50 IU/kg twice daily in a 12-month study. (A) Condensed neurons with both eosinophilic cytoplasm and nuclei (arrows) are present in the medial part of the CA-1 sector and starting at the subiculum that appears predominantly normal. (B) Necrosis is accompanied by vacuolation of the neuropil. Note the abrupt transition from normal to necrotic neurons. H&E, original objectives 6.3× (A), 25× (B).
Figure 7. Dentate gyrus of a rat treated with an insulin analogue at 50 IU/kg twice daily in a 12-month study. Neuronal necrosis is confined to the crest of the dentate gyrus (arrows). Note that the abrupt transition from normal to necrotic neurons (A, B). H&E, original objectives 16× (A), 25× (B).
In the hippocampus, these findings could be clearly distinguished from the single so-called dark neurons present also in control rats and intermingled with the majority of normal, vesicular, and faintly stained neurons (Figure 8). This was different for the cortex, were potential cortical findings occurred infrequently, and appeared to affect only individual or groups of neurons in the superficial layers, and which were difficult to discriminate from “dark neurons” that have to be considered as histological neuronal artifacts (Jortner 2006). Brain stem, basal ganglia, and cerebellum were free from lesions in our toxicity studies with insulins/insulin analogues.
Figure 8. Hippocampus of a control rat: faintly stained neurons with large vesicular and subtle basophilic stained nuclei are intermingled with few dark and shrunken cells suggesting artifacts known as “dark neurons” (arrow). H&E, original objectives 25×.
The findings described above were in overall accordance with those mentioned in the literature, in particular by Auer and his colleagues (1986 and 2004) but also by Mohseni (2001). Hypoglycemia is known to induce neuronal necrosis in cerebral cortex (Agardh et al. 1980; Auer et al. 1985a), hippocampus, and caudoputamen (Kalimo et al. 1985). A distinct lesion pattern is described for the hippocampus (Auer et al. 1984; Auer et al. 1985b; Bree et al. 2009) involving CA-1 and the crest of the gyrus dentatus specifically. The lesions did not necessarily occur bilaterally or symmetrically. The usual development of hypoglycemic lesions in the cortex and hippocampus, associated with a relative resistance of the cerebellum and brain stem, facilitates the differentiation of hypoglycemic findings from those of ischemia-induced lesions (Auer 2004).
In contrast to the literature, where the CA-3 is reported to be relatively resistant to hypoglycemia, this sector was involved occasionally in our rat studies. In addition, under conditions of severe and sustained hypoglycemia, the entire hippocampus became necrotic, but with a severity gradient from CA-1 or the dentate gyrus crest to the other compartments still being present.
Similar microscopic findings have been observed in dogs; however, these were observed less frequently and consistently. Also in the literature, the hippocampus and the cerebral cortex have been described as sites of predilection for hypoglycemic lesions in dogs (Shimada et al. 2000) and cats (Okada et al. 1992).
In our repeated dose toxicity studies, cerebral lesions were exclusively seen in rats and dogs that died or were euthanized prematurely due to clinical observations suggestive (e.g., trembling, convulsions) or indicative (glucose levels) of severe hypoglycemia (Table 1). This may explain why the cerebral histopathological findings were confined to acute necrosis without any accompanying inflammatory reaction. Moreover, it suggested that the brain lesions were not compatible with the survival of the animals because findings have never been observed in animals that reached the end of the study. Subsequently, subacute or chronic stages of brain lesions could not develop. However, a disadvantage was that it was impossible to assess the reversibility of these cerebral findings. Reports in the literature on the reversibility of hypoglycemic brain findings are controversial, but the reversibility is clearly related to severity and duration of hypoglycemia. It should also be noted, however, that the potential of reversibility depends on the tissue compartment affected (Agardh et al. 1980). Nevertheless, it is evident both from the literature (Kalimo et al. 1985) and based upon our own common knowledge from general pathology that no regeneration in the CNS will be possible once necrosis has occurred.
Interestingly, the brain and also the peripheral nervous system (PNS) has been not described as a target organ for recombinant insulins or insulin analogues by other companies (e.g., lispro, aspart, detemir, and degludec) despite the presence of clinical symptoms and unscheduled deaths due to hypoglycemia which have been reported in rodents and dogs within the approval files available on the European Medicines Agency (EMA) and Food and Drug Administration (FDA) home pages. For the animals that survived the reported clinical hypoglycemia, the duration of hypoglycemia, the reduction of the doses used, discontinuation of dosing, or therapeutic intervention to avoid premature deaths could be explanations for this. However, a question remains as to why the unscheduled deaths with preceding hypoglycemia did not show any microscopic changes in the PNS. The pathogenesis of hypoglycemic brain injury is not simply induced by energy deficiency. The presence of decreased glycolysis and subsequent lack of acetate leads to an accumulation of oxalacetate that in turn drives its transamination to aspartate which is then released into the cerebrospinal fluid. Excess of aspartate mediates excitatory and neurotoxic effects resulting in neuronal damage. In addition, intracellular alkalosis due to protein catabolism contributes to this process (Auer and Siesjö 1993; Auer 2004). The release of excitotoxic aspartate into the cerebrospinal fluid may also explain the predilection sites of hippocampus and dentate gyrus that are bathed by the cerebrospinal fluid.

PNS findings related to suprapharmacologic insulin levels

Peripheral neuropathy (PN) in animals under hypoglycemic conditions has been rarely reported in the literature. After decades of development of insulins/insulin analogues, we have observed this entity only during the development program of a long-acting insulin. It was seen in both a 2-week- and a 1-month study in rats and in a 1-month study in 1 dog only and was induced by a long-acting insulin analogue later discontinued due to lack of clinical efficacy. The insulin analogue possessed an extraordinarily long pharmacokinetic/dynamic profile. Microscopically, this unique finding in sciatic nerves showed microscopic features that were compatible with axonal degeneration of the Wallerian type. At low magnification, increased vacuolation was present, and at higher magnifications, vacuoles were filled with condensed eosinophilic material (Figure 9). These eosinophilic globules are known as digestion chambers or ovoid formations (Ikegami et al. 2000; Ozaki et al. 2010) and are thought to be disintegrated and homogenized myelin sheets. Digestion chambers are considered to be a hallmark of PN of the Wallerian type. Occasionally, a subtle inflammatory response consisting of mononuclear cells was present. In most rats affected, degeneration of the sciatic nerves was associated with degenerative changes in thigh muscles considered to be secondary to paresis. These findings were not reversible after the 2-week recovery period.
Figure 9. Sciatic nerve of a rat treated with an insulin analogue at 200 IU/kg in a 2-week study: evidence of increased vacuolation. Vacuoles contain frequently condensed eosinophilic globules—digestion chambers (arrows). H&E, original objectives 25×.
According to the literature on hypoglycemia research, peripheral nerves are only affected in cases of severe (<1.5 mmol/L) and sustained (≥12 hr) hypoglycemia (Yasaki and Dyck 1990, 1991). This corresponds exactly to our unique observation of hypoglycemic PN seen after treatment with an insulin analogue that displayed supralong-acting pharmacodynamic/kinetic properties. These microscopic features induced by a supralong-acting insulin were in accordance with hypoglycemia-related PN reported in the literature for rats (Sima, Zhang, and Greene 1989; Mohseni 2000; Tabata 2000), mice (Tabata 2000), and dogs (Moore et al. 2002). Recently, PN has also been reported in rats using the long-acting insulin analogue Abasria, a biosimilar of insulin glargine (EMA 2014) but not for other insulin analogues, in particular the long-acting insulins approved to date.
The mechanism of hypoglycemic PN is different from that of the CNS because the PNS is able to use other energy sources under hypoglycemic conditions. In contrast to the brain, disturbed blood supply is thought to be involved in the pathogenesis of PN (Eaton and Tesfaye 2003). Hypoglycemia-related lesions were detected in endoneural microvessels of sciatic nerves in rats (Ohshima and Nukada, 2002). Both axons and myelin sheaths were affected. Microscopic features of hypoglycemic neuropathy resembled the Wallerian type of degeneration (Sima, Zhang, and Greene 1989; Mohseni and Hildebrand 1998; Sugimoto et al. 2003; Ozaki et al. 2010). Motor fibers are more susceptible to hypoglycemia-related lesions (Mohseni 2000), whereas sensory fibers are usually affected under hyperglycemic (diabetic) conditions (Sima, Zhang, and Greene 1989). Generally, nerve trunks appear to be more prone to lesions than nerve roots, and within the spinal roots, ventral roots seemed to be the most sensitive. Unfortunately, reversibility of PN has not been tested in the studies published so far.
Overall, in our toxicity studies with insulin/insulin analogues, the nervous system has been more frequently observed as a target organ in rats rather than dogs. One explanation might be that dogs were usually given lower doses based on their supposed higher sensitivity in terms of clinical hypoglycemia (see Table 1). Furthermore, based on the more evident clinical signs of hypoglycemia in dogs, it could be speculated that the decision for dose reduction, using both therapeutic intervention and premature euthanasia, may have been performed earlier. Nevertheless, a scientific explanation for the species-specific sensitivity difference has so far not been elucidated.

Findings in pancreatic Langerhans islets related to suprapharmacologic insulin levels

Islet cell findings after insulin treatment have been rarely published. We have seen islet findings in a 1-month toxicity study in rats with the extraordinarily long-acting insulin analogue that also induced the PN. In control rats, islet cells showed faintly staining homogeneous cytoplasm, and there was no specific distribution pattern visible (Figure 10A). A completely different picture was present in rats after repeated treatment for 1 month with the insulin analogue. Cells at the center of islets were eosinophilic and shrunken and showed dark, condensed nuclei, whereas the larger peripheral cells seemed sometimes to be larger with prominent vesicular and slightly basophilic nuclei, although in many cases, the nuclear density appeared also to be increased in this area when compared to controls. The changes seen in the central area were indicative of an atrophic effect, and in the periphery, the changes were considered to be suggestive of possible hypertrophic effects (Figure 10B). The distribution pattern described, corresponds exactly to the arrangement of insulin producing β cells forming the islet core, whereas glucagon producing α cells are confined to the margin (Wieczorek, Pospischil, and Perentes 1998; Germann et al. 1999) and was further confirmed by immunohistochemical insulin/glucagon double staining (Figure 10C).
Figure 10. Langerhans islets of rats treated with an insulin analogue at 50 IU/kg in a 1-month study with a 2-week recovery period. (A) Langerhans islets of control rats: homogenous appearance and distribution of faintly stained, vacuolated cells. (B) Langerhans islets of a rat treated with an insulin analogue at 50 IU/kg in a 1-month study: islet cells in the core with shrunken, eosinophilic cytoplasm and dark, condensed nuclei are suggestive of atrophy. Faintly stained large marginal islet cells with prominent vesicular nuclei are suggestive of hypertrophy. This pattern corresponds to the distribution of β and α cells in pancreatic islets of rats. (C) Double staining of β cell (red) and α cell (brown) indicating insulin- and glucagon-secreting cells. (D) Langerhans islet of a rat after a 2-week recovery period: the islet cells appear faintly stained, vacuolated, and overall homogenous as in control. There is no longer a difference evident between core and marginal islet cells. H&E, original objectives 40× (A, B, D); Immunohistochemistry (C) with polymer peroxidase (diaminobenzidine) and polymer alkaline phosphatase (fast red), original objective 40×.
With the aldehyde-thionin staining used in the past, β cell degranulation could be frequently demonstrated in both rats and dogs after insulin treatment without evidence of atrophy of islet core cells in H&E-stained slides. It was also interpreted as a kind of disuse atrophy.
After a 2-week recovery period, there was usually no evident difference between the central islet cells and those of the margin indicating probable reversibility. Both cell types displayed faintly stained cytoplasm and nuclei (Figure 10D). The more vesicular appearance of cells and the reduced nuclear density in the central compartment, seen in individual islets, may be suggestive of minimal hypertrophy of these cells.
To our knowledge, these findings in the endocrine pancreas have not been extensively described in scientific literature. We found only a single paper describing the effects of hyperglycemia on pancreatic islets in rats (Clark et al. 1982), where a decrease in the β-cell area was mentioned secondary to hypoglycemic conditions.
These findings in the pancreatic islets of Langerhans should be regarded as adaptive and/or compensatory responses rather than direct effects of insulin/insulin analogues. Exogenous, excessive insulin delivery to normoglycemic rats interferes with the endogenous insulin release of islet cells. It results in a kind of disuse atrophy of β cells which form the core of the islets (Kracht 1958). Comparable islet cell atrophy and interpretation were described for the long-acting insulin analogue Abasria, newly Abasaglar (EMA, 2014) in a repeated toxicity study in rats. It was considered to be consistent with a negative feedback mechanism in hyperinsulinemic animals and a subsequent reduction in endogenous insulin production. In our case, glucagon producing α cells at the margin of pancreatic islets became hypertrophic/hyperplastic in counteracting the exogenous insulin surcharge. After withdrawal of exogenous insulin delivery, the β cells regained their functional activity, and the α-cells are no longer stimulated. Thus, the microscopic differences seen between central and marginal cells disappeared after the treatment-free period. The hypertrophic stimulus on central cells, which is seen occasionally, may thus reflect a compensatory response.

Miscellaneous findings

In a 12-month toxicity study in rats with insulin glulisine and 24-month carcinogenicity studies both in rats and mice with insulin glargine, respectively, treatment-related malignant fibrous histiocytoma (MFH) was diagnosed in male and, occasionally, in female rats and mice (Greaves and Faccini 1981; Ward et al. 1981) of most groups, including the vehicle control group, at the sc administration site on a background of chronic irritation and inflammation (Stammberger et al. 2002; European Medicines Evaluation Agency [EMEA], 2005). Administration-site MFH is a well-known finding and has been reported in rodent long-term studies with drugs, nongenotoxic chemicals, plant extracts, and inert materials when administered or implanted sc (Bartholomew, Kreeger, and Morton 2014; Thomas et al. 1977). Chronic irritation and inflammation were considered to constitute the key etiologic trigger of administration-site MFH (Thomas et al. 1977; Stammberger et al. 2002; Bartholomew, Kreeger, and Morton 2014). Interestingly, administration-site MFH was seen in preclinical rodent studies with a number of other marketed drugs (Bartholomew, Kreeger, and Morton 2014). Even after decades of clinical use of insulin and insulin analogues, no reports of administration-site MFH or other injection-site neoplasia in diabetic patients were identified in the available literature. Therefore, administration-site MFH in rodent long-term studies with insulin glargine and insulin glulisine was considered not to be relevant for humans nor mechanism related.
There were other sporadic observations in laboratory animals related to insulin. Testicular findings, seen in rats treated with the fast-acting insulin analogue aspart (FDA, 2000), were also discussed as being secondary to hypoglycemia. Hypertrophy of rodent parotid and submandibular glands related to insulin treatment has been reported but solely by Wang, Purushotham, and Humphreys-Beher (1994).
These observations, however, lacked consistency and reproducibility across different products and moreover were never proven to be relevant for humans. Therefore, they were beyond the scope of this review which has focused on classical target organs of insulin treatment with potential relevance for humans.

Utility and need for preclinical safety evaluation in diabetic animal models

A relatively wide spectrum of animal models for diabetes is available (see reviews from King 2012; Sasase et al. 2013). Based on the conclusion that all target organ toxicity of the insulins and insulin analogues identified so far is related to exaggerated pharmacology and hypoglycemia, why not evaluate insulin therapy in these hyperglycemic animal models of diabetes? There has been to date no published or documented experience (scientific papers or FDA/EMA approval packages) on the use of diabetic animal models in regulatory toxicity studies with insulins.
The fact that hypoglycemia can be thought of as dose limiting could speak in favor of the use diabetic animals. However, according to our experience and knowledge, there has been no difficulty in achieving favorable safety margins and exposure ratios for insulins and insulin analogues using normoglycemic animals in toxicity testing.
The position paper of Morgan et al. (2013) has outlined the benefits and limitations of animal models of human diseases in nonclinical safety assessment. Based upon the benefit limitation balance, the main recommendation of the paper was that disease models in safety testing should be reserved for the elucidation of toxicity mechanisms, in particular in the context of firstly, unexpected clinical toxicity not found in conventional preclinical studies; and secondly, differentiation of on-target toxicity from off-target toxicity. The recent investigation of unusual pathology related to a hypoglycemic compound (e.g., findings in myocardium and skeletal muscles) by using the Zucker diabetic fatty rat model (Tirmenstein et al. 2015) provides an example for the second case.
However, based on our experience and knowledge, unknown mechanisms of toxicity with unfavorable risk–benefit ratios have not been an issue in the preclinical development of insulins and insulin analogues, and therefore, no clear scientific needs have arisen to use specific animal models. The target organs are well known and characterized and exclusively related to hypoglycemia (on-target toxicity) without any differences between regular insulins and insulin analogues.

Regulatory aspects of the preclinical mitogenicity/carcinogenicity testing strategy for recombinant insulins and their analogues

The International Conference on Harmonization (ICH) has published the ICH S6 (EMEA 1997) guideline specific to the preclinical development of biopharmaceuticals.
Overall, the ICH S6 is applicable for insulins, they being peptide hormones and also for their biosynthetic modifications. In contrast to chemical molecules, classical genotoxicity is expected to be absent for biopharmaceuticals based on the inability of direct DNA interaction and damage. Thus, chemical carcinogenicity has not been expected for peptides, and standard carcinogenicity bioassays are generally considered as inappropriate for biopharmaceuticals by the ICH S6. However, mitogenicity and the potential for tumor promotion is a considerable safety concern for growth promoting hormones (see review by Heidel and Page 2008) and for insulins in particular as already described above.
The approaches to preclinical testing of mitogenicity and carcinogenicity were initially different among companies but became more harmonized over time and have been further facilitated by upcoming regulatory guidance.
The first recombinant (biosynthetic) human insulin was already available for patients worldwide in 1982. Based upon the homology to endogenous human insulin in terms of structure and pharmacology, chronic preclinical toxicity studies, in particular those for mitogenicity and carcinogenicity evaluation were not required for the first and the subsequent recombinant human insulins.
As our understanding of the potential growth promoting effects of insulin increased, the scientific and regulatory concern for potential tumor promoting effects of recombinant insulins likewise increased. This, presumably, was initially driven by the report of Dideriksen, Jorgensen, and Drejer (1992) on the tumorigenic properties of the insulin analogue B12 Asp in rats, but more importantly on the growing molecular knowledge on the causal relationship between mitogenicity and the insulin and insulin-like growth factor (IGF) receptors. Therefore, attempts have been undertaken in the overall preclinical evaluation to study these effects more closely. Interestingly, the approaches used were diverse and have changed over time.
For the first insulin analogue, the fast-acting insulin lispro (approved 1996), the preclinical assessment of mitogenicity was focused on the in vitro evaluation of binding affinity to the insulin and IGF-1 receptors and growth effects on human mammary epithelial cells compared to regular human insulin and the use of insulin B12 Asp as a positive control. Twelve-month toxicity studies were conducted in rats and dogs (FDA, 1996; Zimmermann and Truex 1997; Llewelyn, Slieker, and Zimmermann 1998). Insulin aspart was approved 3 to 4 years later, and the preclinical mitogenicity and carcinogenicity evaluation (FDA, 2000) was comparable to that of lispro (binding affinity and mitogenicity in vitro and 12-month studies in rats and dogs). For insulin glargine, the first long-acting analogue available for patients since 2000, the approach was different. It was evaluated in classical 2-year carcinogenicity studies in rats and mice (Stammberger et al. 2002), in addition to an in vitro mitogenicity testing battery comprising comparative insulin and IGF-1 receptor affinity and thymidine incorporation assays (FDA, 2000), and there was no treatment-related carcinogenicity.
Considering the increasing scientific evidence and resulting regulatory need, the EMEA published the “Points to consider document on the non-clinical assessment of the carcinogenic potential of insulin analogues” in 2001 (pp. 1–5).
Insulin glulisine, approved in 2004, was the first analogue that was preclinically evaluated in compliance with the EMEA “points to consider” paper. Mitogenic potential was evaluated in vivo, in addition to the in vitro testing of the affinity to the insulin and IGF-1 receptor (human osteosarcoma cell line B10), activation of insulin receptor substrates (human myoblasts or rat cardiomyocytes), and DNA synthesis by 3H thymidine incorporation (human breast cell line MCF10) compared to regular human insulin and partially to insulin B12 Asp as a positive control. In vivo cell proliferation was measured in mammary glands of female rats sensitive to hyperplasia/neoplasia and Ki-67 was used as a proliferation marker. This proliferation analysis, which was incorporated into the 6- and 12-month rat toxicity studies, did not indicate any increased mitogenicity as compared to regular human insulin (Stammberger et al. 2006). This in vivo proliferation analysis was specifically requested by a national health authority.
For the long-acting insulin detemir that was approved shortly after, the in vitro evaluations were comparable to insulin glulisine but not the in vivo approach for mitogenicity and carcinogenicity (FDA, 2005). At present, the ultra-long-acting insulin analogue degludec, approved in Europe in 2012 and currently under FDA review, was the last innovative insulin analogue that went to the market. In compliance with the EMEA points to consider paper, in vitro and in vivo investigations were both conducted in equal measure. Cell proliferation was assessed in rat mammary glands from the 12-month toxicity study, using bromodeoxyuridine (BrdU) immunohistochemistry (EMA 2012).

Evaluation of mitogenic activity of insulin and insulin analogues

It should be emphasized that in addition to the metabolic effects mentioned above, insulins show mitogenic activity. This potential tumor promoting activity is related to the insulin receptor and to the IGF-1 receptor linked to respective receptor kinases (see reviews from Sandow 2009; Hvid et al. 2011; Varewick and Janssen 2012) and is already inherent to endogenous regular insulin. Based on this knowledge, a hypothetical safety concern has been raised that the modified sequence and structure of insulin analogues could be associated with enhanced mitogenicity and tumor promotion. Therefore, the need for the preclinical assessment of this mitogenicity/carcinogenicity potential has been the most challenging issue in the preclinical testing of the insulin analogues.
Several approaches have been used in the history of insulin development to address this hypothetical concern as described above.
A principal strategy has been the evaluation of immunohistochemical proliferation markers in regulatory toxicity studies focused on organs with high spontaneous tumor incidences, such as the mammary gland. This approach is scientifically based on the consideration that increased mitogenic potency of modified insulins may promote initiated preformed/preneoplastic cell clones not visible in H&E-stained slides. Mammary gland tumors are among the most prevalent tumors in female rats (Sher, Jensen, and Bokelmann 1982; Brix et al. 2005; Giknis and Clifford 2013). A further basis for this strategy is interestingly a single report that the incidence of benign and malignant mammary tumors was increased in female rats at 100- to 200-fold the therapeutic dose of insulin analogues (Dideriksen, Jorgensen, and Drejer 1992), and the rat mammary gland was shown to express functional insulin and IGF-1 receptors (Hvid et al. 2011). Although the publication by Dideriksen, Jorgensen, and Drejer (1992) report has remained the only case of tumorigenic potential of newly engineered insulin analogues, the evaluation of proliferation markers in preclinical toxicity studies was recommended by a specific points to consider paper (EMEA, 2001).
There are few publications describing the measurement of proliferative activity as an attempt to assess carcinogenic potential of pharmaceuticals preclinically, in particular, in the mammary gland (Stammberger et al. 2006; Christov et al. 2007), although there are a multitude of publications describing different models for the evaluation of initiation/promotion based upon the measurement of DNA synthesis, mitosis, formation of preneoplastic foci, and potential progression of preneoplastic to neoplastic lesions in different organ systems (Pitot and Sirica 1980; Ito et al. 1992; Shirai 1997; Grasl-Kraupp et al. 2000; Ittrich et al. 2003; Tsuda et al. 2003; Grassi et al. 2011; Kushida et al. 2011). Based on our own experience, the preclinical testing of the mitogenic potential is the most challenging aspect in the safety assessment of insulins/insulin analogues because all the other findings due to exaggerated pharmacodynamics are well known today, and it is not difficult to obtain a therapeutic window for the treatment of diabetic humans based upon the use of normoglycemic/nondiabetic animals.
For the assessment of putative mitogenic activity in mammary glands, several technical and scientific aspects have to be taken into account. Among the proliferation markers, Ki-67 is widely accepted and used for the evaluation of histological slides. A good correlation with BrdU has been proven for many species, including the rat (Muskhelishvili et al. 2003) and in particular for the mammary gland (Hvid et al. 2012). In addition, the in vivo injection and further shortcomings of BrdU may be avoided, and therefore, Ki-67 has been recommended as the first-choice marker in rodents (Birner et al. 2001). According to our experience, distinct and reproducible nuclear staining in paraffin sections can be achieved (Figure 11) with a defined formol fixation not longer than 48 hr and after antigen retrieval with microwave/citrate buffer using the MIB-5 antibody (Dako, Hamburg, Germany).
Figure 11. Detection of cell proliferation in the mammary gland using Ki-67 immunohistochemistry aimed on the assessment of the mitogenic potential of insulin analogues. Positive cells are stained red. MIB-5 antibody, alkaline phosphatase/fast red, and counterstain hemalaun; original objectives 18×.
For the mammary gland, it is well known that the proliferative activity is dependent on the hormonal milieu (Russo and Russo 1994) and the estrus cycle stage (Schedin, Mitrenga, and Kaeck 2000; Hvid et al. 2012). Therefore, the rat mammary glands to be evaluated need to be staged for their estrus cycle, mainly assessed on the vaginal histology (Davis, Travlos, and Mc Shane 2001). It is clear that for the counting/evaluation strategy, consistency and representativeness in sampling are needed. In our approach, samples were removed from the thoracic and abdominal portions of mammary glands, and Ki-67-immunostained paraffin sections were prepared.
The evaluation of cell proliferation in vivo was initially confined to development stages of compounds close to submission (12-month studies), but the current approach is to include the in vivo evaluation of mammary gland proliferation already into the 1-month rat studies.
An image analysis system/software tool (Figure 12) was developed, tested, and validated in house for the detection/calculation of proliferation indices in the mammary gland tissue of rats. To ensure robustness and statistical power, at least 1,500 epithelial cells of alveolar and ductular structures per animal were counted in randomly chosen fields. Among the counted cells, the number of Ki-67 positive cells was recorded separately. Only cells with diffuse or stippled red pigment over the nuclei were counted as positive. The proliferative index was calculated as the ratio of stained to total mammary gland cells. Estrous cycle adjusted subgroups from insulin/insulin analogue treated rats were compared to the control group by calculating mean, standard deviation, and median. The statistical significance was tested by using the Kruskall–Wallis 2-sample test.
Figure 12. Quantification of Ki-67 cell proliferation in the mammary gland epithelium with an “in-house” developed image analysis system. Top, left—Ki-67 positive cells (red) and Ki-67 negative cells (green). Top, right—total number of epithelial cells. Bottom, left—proliferating cells (Ki-67 positive).
For all insulin analogues engineered and tested so far by Sanofi, no increased mitogenic or carcinogenic potential compared to regular insulins was demonstrated in preclinical studies, in particular for insulin glargine (Stammberger et al. 2002) and insulin glulisine (Stammberger et al. 2006) approved and on the market. Also other insulin analogues such as insulin lispro, insulin detemir, insulin aspart have not been displayed increased proliferative potential relative to regular human insulin (see review by Vahle et al. 2010).
Obviously, the regulatory environment has evolved to be in favor of investigating the mitogenic potential in vitro and in vivo instead of running classical 2-year carcinogenicity studies.
In summary, for all the insulin analogues available for patients to date and which are cited above, the preclinical safety evaluation did not show any evidence of an increased mitogenic and carcinogenic potential when compared to regular human insulin.

Relevance for humans

The findings described for target organs in the nervous system do not represent species-specific toxic effects in animals. They are relevant for humans because they are directly or indirectly related to exaggerated pharmacodynamics of the compounds resulting in hypoglycemia. Hypoglycemia is defined in man as typical symptoms characterized by plasma glucose concentration ≤3.9 mmol/L (ADA, 2005).
Observations of hypoglycemic brain damage in humans originate interestingly from psychiatry. Sakel’s therapy (1937) of schizophrenia and drug addiction was based on a desired transient hypoglycemic coma. Earlier reports are derived from patients with insulinoma. Today, cases are reported from medication errors (excess or ill-timed insulin administration) as well as from homicide (Haibach, Dix, and Shah 1987; Hood et al. 1986; Levy et al. 1985; Missliwetz 1994) and suicide attempts (Lutz et al. 1997). Severe hypoglycemia, which occurs in the setting of excess or ill-timed insulin administration, has been shown to cause brain damage.
In humans, predilection sites of hypoglycemia in the CNS are similar to animals. As in rats, hippocampus, cerebral cortex, and basal ganglia are usually affected, and the cerebellum is always spared (Kalimo and Olsen 1980; Auer 1986; Fujioka et al. 1997; Bree et al. 2009). Mechanism and pathology of hypoglycemic brain injury in humans were comprehensively investigated and described (Auer 1986; Auer et al. 1989; Auer 2004).
The few reports available for PN in humans are predominantly caused by insulinomas. Similar to rats, axonal degeneration and demyelinization were described. Despite the possible complex etiology involved, hypoglycemia is thought to be the main cause of this type of neuropathy (Eaton and Tesfaye 2003). Adaptive findings in pancreatic islets are related to the exogenous insulin supply to rats with normal endogenous insulin secretion and are, therefore, without relevance for diabetic patients.
In conclusion, it should be highlighted that the findings in the nervous system described here were induced in nondiabetic normoglycemic rats and dogs, in contrast to insulin-deficient, hyperglycemic diabetic patients. Consequently, it is usually not difficult to achieve a therapeutic window for the treatment of diabetic patients for newly developed insulins/insulin analogues.
The more challenging aspect of modern insulin development is evaluation of increased mitogenicity and potential tumor promotion and translating these nonclinical data to an effective human risk assessment. This hypothetical issue has been raised by recent, conflicting, retrospective observational studies, or meta-analyses suggesting a carcinogenic potential of insulin analogues, in particular insulin glargine (Colhoun et al. 2009; Currie, Poole, and Gale 2009; Hemkens et al. 2009; Jonasson et al. 2009; Rosenstock et al. 2009; Du et al. 2012; Ruiter et al. 2012) accompanied by an editorial (Johnson and Gale, 2010). Overall, these studies were considered as inconclusive and the European health authority (EMA, 2013) came to the conclusion that overall the data did not indicate an increased risk of cancer with insulin glargine.
Indeed, experimental evidence indicated that the proliferative effect in vitro of insulin glargine was not much different from that of regular human insulin or other insulin analogues and the rapid enzymatic conversion precluded sustained activation of the IGF-1 receptor signaling pathway (Ciaraldi and Sasaoka 2011; ter Braak et al. 2014). Moreover, pharmacokinetic data from clinical trials have shown that insulin glargine is rapidly converted into its metabolites M1 and M2 in both type 1 (Bolli et al. 2012) and 2 diabetic patients (Lucidi, Porcellati, and Rossetti 2012). The metabolic activity of M1 and M2 is lower and their mitogenic activity similar to regular insulin (Sommerfeld et al. 2010). Therefore, it has been concluded that the in vitro findings of increased mitogenicity are not translatable to the clinical setting in humans (Owens 2012; Tennagels and Werner 2013).
In addition, besides the mitogenicity hypothesis, the hyperglycemia hypothesis (providing tumor cells with more fuel) was also discussed (Johnson and Gale 2010) resulting in the conclusion that the link between insulin treatment and cancer risk might be rather driven by the high glucose levels than by any direct effect of insulin. This hypothesis was supported by the clinical trial conducted by Yang et al. (2010) indicating an increased cancer risk in diabetic patients lacking insulin treatment, whereas the insulin use was associated with reduced risk.

Conclusions

The toxicological profile of insulins/insulin analogues in preclinical studies with laboratory animals is exclusively due to exaggerated pharmacodynamic activity induced by suprapharmacological dose levels. The usual target organ is the CNS with a specific microscopic pattern in the hippocampus and dentate gyrus. To our knowledge, findings in the PNS have been rarely reported in preclinical toxicity studies. Findings in the islets of Langerhans in nondiabetic animals have to be considered adaptive in nature and not directly related to insulins/insulin analogues.
Insulin analogues displaying modifications of peptide sequence and structure are aimed on the tailoring of pharmacodynamics/kinetics to the physiological pattern of glucose release. However, for these modifications, there is so far no proof that they may result in an altered toxicological profile.
The CNS and PNS findings obtained in preclinical studies are relevant for human patients. However, it is not difficult to achieve a safety margin and exposure ratio for patients suffering from diabetes, because the microscopic findings were induced in nondiabetic, normoglycemic laboratory animals, being naturally more sensitive to the hypoglycemic effects of exogenous insulin.
Despite exaggerated pharmacodynamic activity being thought of as a dose-limiting event, there has been no scientific need to date for the use of diabetic animal models in toxicity studies, because toxicity mechanisms with resulting unfavorable risk:benefit ratios have not been experienced with insulin analogues. More challenging is the evaluation of the mitogenic potential related to the insulin and IGF-1 receptors, which is already inherent to regular insulins. Regulatory guidance is available for the preclinical mitogenicity/carcinogenicity testing. In addition to the in vitro testing battery, the in vivo evaluation has been usually performed by the incorporation of proliferation marker analysis into regulatory toxicity studies in rats. The regulatory environment has evolved such that it is now favored to investigate proliferation using specific in vitro and in vivo studies instead of running classical 2-year carcinogenicity studies.
For all commercially available recombinant insulins and their analogues on the marked and additional compounds tested preclinically by Sanofi so far, there has been no evidence of enhanced mitogenicity beyond that already innate to endogenous insulin.

Acknowledgments

The authors would like to thank the members of the Regulatory Forum committee for suggestions and fruitful discussions, in particular Armando Irizarry Rovira to adapt the article to the needs of the Regulatory Forum. We would like to thank Juergen Korus (Histology) and Barbara Gennat (Immunohistochemistry) from Sanofi, Frankfurt, for their excellent technical work.

Authors’ Note

This paper was initially submitted to Toxicologic Pathology as a scientific review on the toxicological pathology profile of insulins and insulin analogues with only little attention on regulatory aspects. Upon encouragement of the Regulatory Forum committee and in close interaction with them, the review was extended with chapters on regulatory aspects. The considerations on regulatory aspects reflect the personal opinion and experience of the authors and do not necessarily constitute an official position of the company Sanofi.

Declaration of Conflicting Interests

The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

Funding

The author(s) received no financial support for the research, authorship, and/or publication of this article.

References

ADA (American Diabetes Association)—Workgroup on hypoglycemia. (2005). Defining and reporting hypoglycemia in diabetes. Diabetes Care 28, 1245–49.
Agardh C.-D., Kalimo H., Olsson Y., Siesjö B. K. (1980). Hypoglycemic brain injury. I. Metabolic and light microscopic findings in rat cerebral cortex during profound insulin-induced hypoglycemia and in the recovery period following glucose administration. Acta Neuropathol 50, 31–41.
Auer R. N. (1986). Progress review: Hypoglycemic brain damage. Stroke 17, 699–708.
Auer R. N. (2004). Hypoglycemic brain damage. Metab Brain Dis 19, 169–75.
Auer R. N., Hugh J., Cosgrove E., Curry B. (1989). Neuropathologic findings in three cases of profound hypoglycemia. Clin Neuropathol 8, 63–68.
Auer R. N., Kalimo H., Olsson Y., Siesjö B. K. (1985a). The temporal evolution of hypoglycemic brain damage. I. Light- and electron-microscopic findings in the rat cerebral cortex. Acta Neuropathol 67, 13–24.
Auer R. N., Kalimo H., Olsson Y., Siesjö B. K. (1985b). The temporal evolution of hypoglycemic brain damage. II. Light- and electron-microscopic findings in the hippocampal gyrus and the subiculum of the rat. Acta Neuropathol 67, 25–36.
Auer R. N., Siesjö B. K. (1993). Hypoglycemia: Brain neurochemistry and neuropathology. Baillieres Clin Endocrinol Metab 19, 169–75.
Auer R. N., Wieloch T., Olsson Y., Siesjö B. K. (1984). The distribution of hypoglycemic brain damage. Acta Neuropathol 64, 177–91.
Bartholomew P. M., Kreeger J. M., Morton D. (2014). Injection-site malignant fibrous histiocytomas in a pegvisomant carcinogenicity study in SD rats. Toxicol Pathol 42, 1082–91.
Birner P., Ritzi M., Musahl C., Knippers R., Gerdes J., Voigtlaender T., Budka H., Hainfellner J. A. (2001). Technical advance—Immunohistochemical detection of cell growth fraction in formalin-fixed and paraffin-embedded murine tissue. Am J Pathol 158, 1991–96.
Bolli G. B., Hahn A. D., Schmidt R., Eisenblaetter T., Dahmen R., Heise T., Becker R. H. (2012). Plasma exposure to insulin glargine and its metabolites M1 and M2 after subcutaneous injection of therapeutic and supratherapeutic doses of glargine in subjects with type 1 diabetes. Diabetes Care 35, 2626–30.
Bree A. J., Puente E. C., Daphna-Iken D., Fisher S. J. (2009). Diabetes increases brain damage caused by severe hypoglycemia. Am J Physiol Endocrinal Metab 297, 194–201.
Brix A. E., Nyska A., Haseman J. K., Sells D. M., Jokinen M. P., Walker N. J. (2005). Incidence of selected lesions in control female Harlan Sprague-Dawley rats from two-year studies performed by the National Toxicology Program. Toxicol Path 33, 477–83.
Christoph K., Grubbs C. J., Shilkaitis A., Juliana M. M., Lubet R. A. (2007). Short-term modulation of cell proliferation and apoptosis and preventive/therapeutic efficacy of various agents in a mammary cancer model. Clin Cancer Res 13, 5488–96.
Ciaraldi T. P., Sasaoka T. (2011). Review of the in vitro interaction of insulin glargine with the insulin/insulin-like growth factor system: Potential implications for metabolic and mitogenic activities. Horm Metab Res 43, 1–10.
Clark A., Brown E., King T., Vanhegan R. I., Turner R. C. (1982). Islet changes induced by hyperglycemia in rats—Effect of insulin or chlorpropamide therapy. Diabetes 31, 319–25.
Colhoun H. M., and SDRN Epidemiology Group. (2009). Use of insulin glargine and cancer incidence in Scotland: A study from the Scottish Diabetes Research Network Epidemiology Group. Diabetologia 52, 1755–65.
Currie C. J., Poole C. D., Gale E. A. M. (2009). The influence of glucose-lowering therapies on cancer risk in type 2 diabetes. Diabetologia 52, 1766–77.
Davis B. J., Travlos G., Mc Shane T. (2001). Reproductive endocrinology and toxicological pathology over the life span of the female rodent. Toxicol Path 29, 77–83.
Dideriksen L. H., Jorgensen L. N., Drejer K. (1992). Carcinogenic effect on female rats after 12 months administration of the insulin analogue B10 Asp. Diabetes 41, 507.
Du X., Zhang R., Xue Y., Li D., Cai J., Zhou S., Huang Z., Yu R., Liu J. (2012). Insulin glargine and risk of cancer: A meta-analysis. Int J Biol Markers 27, 241–46.
Eaton S. E. M., Tesfaye S. (2003). Insulin, hypoglycemia and peripheral neuropathy. In Textbook of Diabetic Neuropathy (Gries F. A., Cameron N. E., Low P. A., Ziegler D., eds.), pp. 306–9. Thieme, Stuttgart, Germany.
EMA (European Medicines Agency). (2012). European Medicines Agency. Insulin degludec-Tresiba—Committee for Medicinal Products for Human Use (CHMP) assessment report, pp. 11–18. EMA/CPMP/557821/2012, London, September 20, 2012. Accessed August 10, 2015. http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/002498/human_med_001609.jsp&mid=WC0b01ac058001d124.
EMA (European Medicines Agency). (2013). European Medicines Agency. Outcome of review of new safety data on insulin glargine. EMA/329790/2013. European Medicines Agency, London, UK, May 31, 2013.
EMA (European Medicines Agency). (2014). European Medicines Agency. Abasria (now Abasaglar)—Assessment report, p. 20. EMA/CPMP/340840/2014, London, UK, June 26, 2014. Accessed July 21, 2015. http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/002835/human_med_001790.jsp&mid=WC0b01ac058001d124.
EMEA (European Medicines Evaluation Agency). (1997). European Medicines Evaluation Agency. ICH Topic S6. Preclinical safety evaluation of biotechnology-derived pharmaceuticals. CPMP/ICH/302/95. European Medicines Evaluation Agency, London, UK, July 16, 1997.
EMEA (European Medicines Evaluation Agency). (2001). European Medicines Evaluation Agency. Points to consider document on the non-clinical assessment of the carcinogenic potential of insulin analogues. CPMP/SWP/372/01. European Medicines Evaluation Agency, London, UK. November 15, 2001.
EMEA (European Medicines Evaluation Agency). (2005). European Medicines Evaluation Agency. Insulin glulisine-Apidra—European Public Assessment Report (EPAR)—Scientific discussion, p. 12. Accessed August 18, 2015. http://www.ema.europa.eu/ema/index.jsp?curl=pages/medicines/human/medicines/000557/human_med_000648.jsp&mid=WC0b01ac058001d124.
Evans M., Schumm-Draeger P. M., Vora J., King A. B. (2011). A review of modern insulin analogue pharmacokinetic and pharmacodynamic profiles in type 2 diabetes: Improvements and limitations. Diabetes Obes Metab 13, 677–84.
FDA (Food and Drug Administration). (1996). U.S. Food and Drug Administration. Insulin lispro-Humalog. NDA 20-563, Pharmacology Review, pp. 29–30. Accessed August 19, 2015. https://www.pharmapendium.com/fda.do?drugName=Insulin+Lispro+%28Human+Analog%29.
FDA (Food and Drug Administration). (2000). U.S. Food and Drug Administration. Insulin aspart-NovoLog. New Drug Application (NDA) 20-986, Pharmacology Reviews, pp. 65–70. Accessed August 19, 2015. http://www.accessdata.fda.gov/drugsatfda_docs/nda/2000/20-986_NovoLog_pharmr_P4.pdf.
FDA (Food and Drug Administration). (2000). U.S. Food and Drug Administration. Insulin glargine-Lantus. New Drug Application (NDA) 21-081, Pharmacology Reviews, pp. 4–6. Accessed August 16, 2015. http://www.accessdata.fda.gov/drugsatfda_docs/nda/2000/21081_Lantus.cfm.
FDA (Food and Drug Administration). (2005). U.S. Food and Drug Administration. Insulin detemir-Levemir. New Drug Application (NDA) 21-536, Pharmacology Reviews, pp. 21–62. Accessed August 19, 2015. http://www.accessdata.fda.gov/drugsatfda_docs/nda/2005/021-536_LevemirTOC.cfm.
Fujioka M., Okuchi K., Hiramatsu K.-I., Sakaki T., Sakaguchi S., Ishi J. (1997). Specific changes in human brain after hypoglycemic injury. Stroke 28, 584–87.
Germann P. G., Heinrichs M., Püschner H., Hellmann H., Kohler M., Ernst H. (1999). RITA/Registry of Industrial toxicology Animal data: A comparative immunohistochemical study of 77 islet cell carcinomas in Sprague-Dawley and Wistar rats using antibodies against insulin, glucagon, somatostatin and gastrin. Exp Toxicol Pathol 51, 477–487.
Giknis M. L. A., Clifford C. B. (2013). Compilation of Spontaneous Neoplastic Lesions and Survival in Crl: CD (SD) Rats Control Groups (p. 10). Charles River Laboratories. Accessed June 9, 2015. www.criver.com.
Grasl-Kraupp B., Luebeck G., Wagner A., Low-Baselli A., de Gunst M., Waldhor T., Moolgavkar S., Schulte-Hermann R. (2000). Quantitative analysis of tumour initiation in rat liver. Role of cell replication and cell death (apoptosis). Carcinogenesis 21, 1411–21.
Grassi T. F., Marcheson Rodrigues M. A., de Camargo J. L., Barbison L. F. (2011). Evaluation of carcinogenic potential of Diuron in a rat mammary two-stage carcinogenesis model. Toxicol Pathol 39, 486–95.
Greaves P., Faccini J. M. (1981). Spontaneous fibrous histiocytic neoplasms in rats. Br J Cancer 43, 402–11.
Haibach H., Dix J. D., Shah J. H. (1987). Homicide by insulin administration. J Forensic Sci 32, 208–16.
Heidel S. M., Page T. J. (2008). Current Practices in the preclinical safety assessment of peptides. In Preclinical Safety Evaluation of Biopharmaceuticals: A Science Based Approach to Facilitating Clinical Trials (Cavagnaro J. A., ed.), pp. 501–13. Johne Wiley, New York.
Hemkens L. G., Grouven U., Bender R., Guenster C., Gutschmidt S., Selke G. W., Sawicki P. T. (2009). Risk of malignancies in patients with diabetes treated with human insulin or insulin analogues. A cohort study. Diabetologia 52, 1732–44.
Hirsch I. B. (2005). Drug therapy: Insulin analogues. N Engl J Med 352, 174–83.
Hood I., Mirchandani H., Monforte J., Stacer W. (1986). Immunohistochemical demonstration of homicidal insulin injection site. Arch Pathol Lab Med 110, 973–74.
Hvid H., Fels J. J., Rikke K. K., Thorup I., Jensen H. E., Hansen B. F., Oleksiewicz M. B. (2011). In situ phosphorylation of Akt and ERK1/2 in rat mammary gland, colon and liver following treatment with human insulin and IGF-1. Toxicol Pathol 39, 623–40.
Hvid H., Thorup I., Sjoegren I., Oleksiewicz M. B., Jensen H. E. (2012). Mammary gland proliferation in female rats: Effects of the estrous cycle, pseudo-pregnancy and age. Exp Toxicol Pathol 64, 321–32.
Ikegami H., Tabata H., Matsuzawa T., Suzuki H. (2000). The exacerbating effect of insulin-induced hypoglycemia on spontaneous peripheral neuropathy in aged B6C3F1 mice. J Toxicol Sci 25, 137–42.
Ito N., Shirai T., Hasegawa R. (1992). Medium-term bioassays for carcinogens. In International Agency for Research on Cancer (Vainio H., Magee N., McGregor D. B., McMichael A. J., eds.), pp. 353–88. Lyon, France.
Ittrich C., Deml E., Oesterle D., Kuttler K., Mellert W., Brendler-Schwaab S., Enzmann E., Schladt L., Bannasch P., Haertel T., Monnikes O., Schwartz M., Kopp-Schneider A. (2003). Prevalidation of a rat liver foci bioassay (RLFB) based on results from 1600 rats. A study report. Toxicol Pathol 31, 60–79.
Johnson J. A., Gale E. A. M. (2010). Diabetes, insulin use and cancer risk: Are observational studies part of the solution—Or part of the problem? Diabetes 59, 1129–31.
Jonasson J. M., Liung R., Talbaeck M., Haglund B., Gudbjoernsdottir S., Steineck G. (2009). Insulin glargine use and short-term incidence of malignancies—A population-based follow-up study in Sweden. Diabetologia 52, 1745–54.
Jortner B. S. (2006). The return of the dark neuron. A histological artifact complicating contemporary neurotoxicologic evaluation. Neuro Toxicol 27, 628–34.
Kalimo H., Auer R. N., Siesjö B. K. (1985). The temporal evolution of hypoglycemic brain damage. III. Light- and electron-microscopic findings in the rat caudoputamen. Acta Neuropathol 67, 37–50.
Kalimo H., Olsen Y. (1980). Effect of severe hypoglycemia on the human brain—Neuropathologic case reports. Acta Neurol Scand 62, 345–56.
King A. J. F. (2012). The use of animal models in diabetes research. Br J Pharmacol 166, 877–94.
Kracht J. (1958). Morphologische Kriterien zur Beurteilung der Inselzellaktivität. Endokrinologie 36, 146–58.
Kushida M., Kamendulis L. M., Peat T. J., Klaunig E. (2011). Dose-related induction of hepatic preneoplastic lesions by diethylnitrosamine in C57Bl/ mice. Toxicol Pathol 39, 776–86.
Levy W. J., Gardner D., Moseley J., Dix J., Gaede S. E. (1985). Unusual problems for the physician in managing a hospital patient who received a malicious insulin overdose. Neurosurgery 17, 992–96.
Llewelyn J., Slieker L. J., Zimmermann J. L. (1998). Preclinical studies on insulin lispro. Drugs Today 34, 11–21.
Lucidi P., Porcellati F., Rossetti P. (2012). Metabolism of insulin glargine after repeated daily subcutaneous injections in subjects with type 2 diabetes. Diabetes Care 35, 2647–49.
Lutz R., Pedal I., Wetzel C., Mattern R. (1997). Insulin injection sites: Morphology and immunohistochemistry. Forensic Sci Inter 90, 93–101.
Missliwetz J. (1994). Die Mordserie im Krankenhaus Wien-Lainz. Arch Kriminol 194, 1–7.
Mohseni S. (2000). Hypoglycemic neuropathy in diabetic BB/Wor rats treated with insulin implants affects ventral root axons but not dorsal root axons. Acta Neuropathol 100, 415–20.
Mohseni S. (2001). Hypoglycemic neuropathy. Acta Neuropathol 102, 413–21.
Mohseni S., Hildebrand C. (1998). Neuropathy in diabetic BB/Wor rats treated with insulin implants. Acta Neuropathol 96, 144–50.
Moore A. S., Nelson R. W., Henry C. J., Rassnick K. M., Kristal O., Ogilvie K. G., Kintzer P. (2002). Streptozotocin for treatment of pancreatic islet cell tumors in dogs: 17 cases (1989-1999). J Am Vet Med Assoc 221, 811–18.
Morgan S. J., Elangbam C. S., Berens S., Janovitz E., Vitsky A., Zabka T., Conour L. (2013). Use of animal models of human disease for nonclinical safety assessment of novel pharmaceuticals. Toxicol Pathol 41, 508–18.
Muskhelishvili L., Latendresse J. R., Kodell R. L., Henderson E. B. (2003). Evaluation of cell proliferation in rat tissues with BrdU, PCNA, Ki-67(MIB-5) immunohistochemistry and in situ hybridization for histone mRNA. J Histochem Cytochem 51, 1681–88.
Ohshima J., Nukada H. (2002). Hypoglycemic neuropathy: Microvascular changes due to recurrent hypoglycemic episodes in rat sciatic nerve. Brain Res 947, 84–89.
Okada R., Shibutani M., Matsuo T., Tajima T. (1992). Pathological effects of maternal hypoglycemia on fetal development in cats. Acta Pathol Jpn 42, 316–24.
Owens D. R. (2002). New horizons—Alternative routes for insulin therapy. Nat Rev Drug Discov 1, 529–40.
Owens D. R. (2012). Glargine and cancer: Can we now suggest closure? Diabetes Care 35, 2426–28.
Owens D. R., Zinman B., Bolli G. B. (2001). Insulins today and beyond. Lancet 358, 739–46.
Ozaki K., Sano T., Tsuji N., Matsuura T., Narama I. (2010). Insulin-induced hypoglycemic peripheral motor neuropathy in spontaneously diabetic WBN/Kob rats. Comp Med 60, 282–87.
Pitot H. C., Sirica A. E. (1980). The stages of initiation and promotion in hepatocarcinogenesis. Biochim Biophys Acta 605, 191–215.
Rosenstock J., Fonseca V., McGill J. B., Riddle M., Hallé J. P., Hramiak I., Johnson P., Davis M. (2009). Similar risk of malignancies with insulin glargine and neutral protamine Hagedorn (NPH) insulin in patients with type 2 diabetes: Findings from a 5 year randomized, open-label study. Diabetologia 52, 1971–73.
Ruiter R., Vissner L. E., van Herk-Sukel M. P. P., Coeberg J. W. W., Haak H. R., Geelhoed-Duijvestijn P. H., Strauss S. M. J. M., Herings R. M. C., Stricker B. H. Ch. (2012). Risk of cancer in patients on insulin glargine and other insulin analogues in comparison with those on human insulin: Results from a large population-based follow-up study. Diabetologia 55, 51–62.
Russo I. H., Russo J. (1994). Ageing of the mammary gland—Effects of hormones on the development of the young virgin rat. In Pathology of the Aging Rat (Mohr U., Dungworth D. L., Capen C. C., eds.), pp. 450–453. ILSI Press, Washington, DC.
Sakel M. (1937). The methodical use of hypoglycemia in the treatment of psychoses. Am J Psychiatry 94, 111–29.
Sandow J. (2009). Growth effects of insulin and insulin analogues. Arch Physiol Biochem 115, 72–85.
Sasase T., Pezzolesi M. G., Yokoi N., Yamada T., Matsumoto K. (2013). Animal models of diabetes and metabolic disease. J Diabetes Res 2013, 1–2.
Schedin P., Mitrenga T., Kaeck M. (2000). Estrous cycle regulation of mammary epithelial cell proliferation, differentiation and death in Sprague-Dawley rat: A model for investigating the role of estrous cycling in mammary carcinogenesis. J Mammary Gland Biol Neoplasia 5, 211–25.
Sher S. P., Jensen R. D., Bokelmann D. L. (1982). Spontaneous tumors in control F344 and Charles River-CD rats and Charles River CD-1 and B6C3HF1 mice. Toxicol Letters 11, 103–10.
Shimada A., Morita T., Ikeda N., Torii S., Haruna A. (2000). Hypoglycemic brain lesions in a dog with insulinoma. J Comp Path 122, 67–71.
Shirai T. (1997). A medium-term liver bioassay as a rapid in vivo test for carcinogenic potential: A historical review of model development and summary of results from 291 tests. Toxicol Pathol 25, 453–60.
Sima A. A. F., Zhang W.-X., Greene D. A. (1989). Diabetic and hypoglycemic neuropathy—A comparison in the BB rat. Diabetes Res Clin Pract 6, 279–96.
Sommerfeld M. R., Müller G., Tschank G., Seipke G., Habermann P., Kurrle R., Tennagels N. (2010). In vitro metabolic and mitogenic signaling of insulin glargine and its metabolites. PLoS ONE 5, e9540.
Stammberger I., Bube A., Durchfeld-Meyer B., Donaubauer H., Troschau G. (2002). Evaluation of the carcinogenic potential of insulin glargine (Lantus) in rats and mice. Int J Toxicol 21, 171–79.
Stammberger I., Seipke G., Bartels T. (2006). Insulin glulisine—A comprehensive preclinical evaluation. Int J Toxicol 25, 25–33.
Sugimoto K., Baba M., Suda T., Yasujima M., Yagihashi S. (2003). Peripheral neuropathy and microangiopathy in rats with insulinoma: Associated with chronic hyperinsulinemia. Diabetes Metabol Res Rev 19, 392–400.
Tabata H. (2000). Peripheral neuropathy in B6C3F1 mice and SD rats induced by chronic intermittent insulin hypoglycemia. Drug Chem Toxicol 23, 485–96.
Tennagels N., Werner U. (2013). The metabolic and mitogenic properties of basal insulin analogues. Arch Physiol Biochem 119, 1–14.
ter Braak B., Siezen C. L. E., Kannegieter N., Koedoot E., van de Water B., van de Laan J. W. (2014). Classifying the adverse mitogenic mode of action of insulin analogues using a novel mechanism-based genetically engineered human breast cancer cell panel. Arch Toxicol 88, 953–66.
Thomas C., Steinhardt H. J., Küchemann K., Maas D., Riede U. N. (1977). Soft tissue tumors in the rat. Pathogenesis and histopathology. Curr Topic Pathol 64, 129–76.
Tirmenstein M., Horvath J., Graziano M., Mangipudy R., Dorr T., Colman K., Zinker B., Kirby M., Cheng P. T. W., Patrone L., Kozlovsky J., Reilly T. P., Wang V., Janovitz E. (2015). Utilization of the Zucker Diabetic Fatty (ZDF) Rat model for investigating hypoglycemia-related toxicities. Toxicol Pathol 43, 825–37.
Tsuda H., Fukushima S., Wanibuchi H., Morimura K., Nakae D., Imaida K., Tatematsu M., Hirose M., Wakabayashi K., Moore M. A. (2003). Value of GST-P positive preneoplastic hepatic foci in dose-response studies of hepatocarcinogenesis. Evidence for practical thresholds with both genotoxic and nongenotoxic carcinogens. A review of recent work. Toxicol Pathol 31, 80–86.
Vahle J. L., Finch G. L., Heidel S. M., Hovland D. N., Ivens I., Parker S., Ponce R. A., Sachs C., Steigerwald R., Short B., Todd M. D. (2010). Carcinogenicity assessments of biotechnology-derived pharmaceuticals: A review of approved molecules and best practice recommendation. Toxicol Pathol 38, 522–53.
Varewijck A. J., Janssen J. A. M. J. L. (2012). Insulin and its analogues and their affinities for the IGF1 receptor. Endocrine Cancer 19, 63–75.
Wang P. L., Purushotham K. R., Humphreys-Beher M. G. (1994). Effect of chronic insulin administration on mouse parotid and submandibular gland function. Proc Soc Ex Biol Med 205, 353–61.
Ward J. M., Kulwich B. A., Reznik G., Berman J. J. (1981). Malignant fibrous histiocytoma. An unusual neoplasm of soft-tissue origin in the rat that is different from the human counterpart. Arch Pathol Lab Med 105, 313–16.
Wieczorek G., Pospischil A., Perentes E. (1998). A comparative immunohistochemical study of pancreatic islets in laboratory animals (rats, dogs, minipigs, nonhuman primates). Exp Toxicol Pathol 50, 151–72.
Yang X., Ko G. T. C., So X. Y., Ma R. C. W., Yu L. W. L., Kong A. P. S., Zhao H., Chow C. C., Tong P. C. Y., Chan J. C. N. (2010). Association of hyperglycemia and insulin usage with the risk of cancer in type 2 diabetes: The Hong Kong Diabetes Registry. Diabetes 59, 1254–60.
Yasaki S., Dyck P. J. (1990). Duration and severity of hypoglycemia needed to induce neuropathy. Brain Res 531, 8–15.
Yasaki S., Dyck P. J. (1991). Spatial distribution of fiber degeneration in acute hypoglycemic neuropathy in rat. J Neuropathol Exp Neurol 50, 681–92.
Zimmermann J. L., Truex L. L. (1997). 12-Month chronic toxicity study of Ly275585 (human insulin analog) administered subcutaneously to Fischer 344 rats. Int J Toxicol 16, 639–57.

Cite article

Cite article

Cite article

OR

Download to reference manager

If you have citation software installed, you can download article citation data to the citation manager of your choice

Share options

Share

Share this article

Share with email
EMAIL ARTICLE LINK
Share on social media

Share access to this article

Sharing links are not relevant where the article is open access and not available if you do not have a subscription.

For more information view the Sage Journals article sharing page.

Information, rights and permissions

Information

Published In

Article first published online: September 22, 2016
Issue published: October 2016

Keywords

  1. insulin
  2. insulin analogues
  3. hypoglycemia
  4. neuronal necrosis
  5. peripheral neuropathy
  6. Langerhans islets atrophy
  7. mitogenic potential

Rights and permissions

© The Author(s) 2016.
Request permissions for this article.
PubMed: 27663844

Authors

Affiliations

Thomas Bartels
Sanofi, Preclinical Safety, Paris, France
Kerstin Wäse
Sanofi, Preclinical Safety, Frankfurt, Germany
Martin Heinrichs
Consultant in Toxicologic Pathology, Oberursel, Germany
Manuela Stolte
Sanofi, Preclinical Safety, Frankfurt, Germany
Nigel Roome
Consultant in Toxicology and Toxicologic Pathology, Versailles, France
Petra Scherer
Sanofi, Animal Research and Welfare, Frankfurt, Germany
Klaus Lindauer
Sanofi, Frankfurt, Drug Disposition, Germany

Notes

Thomas Bartels, Sanofi, DSAR, Preclinical Safety, 3 digue d’Alfortville, 94140 Alfortville, France. Email: [email protected]
*
This is an opinion article submitted to the Regulatory Forum and does not constitute an official position of the Society of Toxicologic Pathology or the journal Toxicologic Pathology. The views expressed in this article are those of the authors and do not necessarily represent the policies, positions, or opinions of their respective agencies and organizations. The Regulatory Forum is designed to stimulate broad discussions of topics relevant to regulatory issues in Toxicologic Pathology. Readers of Toxicologic Pathology are encouraged to send their thoughts on these articles or ideas for new topics to [email protected].

Authors’ Contribution

Authors contributed to conception or design (TB, KW); data acquisition, analysis, or interpretation (TB, KW, MH, MS, NR, PS, KL); drafting the manuscript (TB); and critically revising the manuscript (TB, KW, MH, MS, NR, PS, KL). All authors gave final approval and agreed to be accountable for all aspects of work in ensuring that questions relating to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Metrics and citations

Metrics

Journals metrics

This article was published in Toxicologic Pathology.

VIEW ALL JOURNAL METRICS

Article usage*

Total views and downloads: 1504

*Article usage tracking started in December 2016


Altmetric

See the impact this article is making through the number of times it’s been read, and the Altmetric Score.
Learn more about the Altmetric Scores



Articles citing this one

Receive email alerts when this article is cited

Web of Science: 2 view articles Opens in new tab

Crossref: 0

  1. Endocrine System
    Go to citation Crossref Google Scholar
  2. Risk Management and Communication: Building Trust and Credibility With...
    Go to citation Crossref Google Scholar
  3. Development and Validation of an Image Analysis System for the Measure...
    Go to citation Crossref Google ScholarPub Med
  4. Nonproliferative and Proliferative Lesions of the Rat and Mouse Endocr...
    Go to citation Crossref Google Scholar

Figures and tables

Figures & Media

Tables

View Options

View options

PDF/ePub

View PDF/ePub

Get access

Access options

If you have access to journal content via a personal subscription, university, library, employer or society, select from the options below:

STP members can access this journal content using society membership credentials.

STP members can access this journal content using society membership credentials.


Alternatively, view purchase options below:

Purchase 24 hour online access to view and download content.

Access journal content via a DeepDyve subscription or find out more about this option.