<<

The growth and energy balance consequences of adolescent abuse

Rose Crossin BSc (Hons)

ORCID: https://orcid.org/0000-0003-1814-1330

Submitted in total fulfilment of the requirements for the degree

Doctor of Philosophy

October 2018

The Florey Department of Neuroscience and

The University of Melbourne

Rose Crossin (737900)

Abstract Inhalant abuse is the intentional inhalation of products containing toluene (e.g. glue or petrol) in order to induce an altered mental state. The primary populations who misuse are adolescents, particularly those in vulnerable or disadvantaged groups, including indigenous peoples. Inhalant abuse has been associated with symptoms such as appetite suppression and impaired weight gain, suggesting that it may have an effect on energy balance. Disruptions to energy balance may have an effect on growth, particularly during periods of rapid growth such as adolescence. However, the energy balance and growth consequences arising from adolescent inhalant abuse have never been fully characterised, and the persistence of these during abstinence remains unknown.

Thus, the aims of this PhD thesis were to:

1. characterise the energy balance and growth consequences of adolescent inhalant abuse; 2. understand how these effects may persist into abstinence, and; 3. elucidate the mechanisms underlying these changes.

Initial exploration of the growth consequences of adolescent inhalant abuse, and the persistence of effects into abstinence, was conducted using a retrospective analysis of data collected from a cohort of Australian Indigenous males who chronically sniffed petrol during adolescence. This investigation was further expanded by conducting a systematic review and meta-analysis on the growth consequences of inhalant abuse (in clinical studies) and toluene exposure (in pre-clinical studies). This included sub-group analysis to identify potential moderators of the effects. Characterisation of the effects of adolescent inhalant abuse, within the context of the energy balance equation, and the testing of potential mechanistic hypotheses, was conducted using an established rodent model of chronic intermittent toluene (CIT) exposure. In the CIT model, male adolescent rats were exposed to toluene with exposure patterns replicating human inhalant abuse, including a period of sustained abstinence.

Adolescent inhalant abuse significantly impaired both body weight and height, which was consistent across the epidemiological study, meta-analysis, and rodent model. These changes persisted into sustained abstinence. Furthermore, using the rodent model, it was determined that adolescent inhalant abuse induces a negative energy balance phenotype, with decreased food intake occurring concurrently with increased energy expenditure at the end of the exposure period. Potential causal hypotheses for the observed growth impairments were tested. Using a pair-fed group, the hypothesis that toluene induced under-nutrition underlies the observed growth impairments was tested. This hypothesis was rejected by the data, with differences between the pair-fed and CIT group related to homeostatic recovery of body weight, skeletal responses to under-nutrition, and glycaemic status indicating different mechanisms are involved in CIT-induced changes to body weight. A further causal hypothesis was identified, positing that adrenal insufficiency may underlie the observed negative energy balance phenotype and growth impairments. This hypothesis was supported by the data, with adrenal histology and endocrine testing revealing results consistent with primary adrenal

1 Rose Crossin (737900) insufficiency, including adrenal hypertrophy, and increased basal adrenocorticotropic hormone (ACTH) concurrent with no increase in basal corticosterone.

In conclusion, these studies revealed that adolescent inhalant abuse causes significant and persistent growth impairments. These have the potential to lead to further adverse health consequences. Body weight suppression has the potential to result in cognitive impairment, skeletal disorders, and increase the risk of an individual developing a metabolic disorder if weight is regained. Additionally, height impairment is associated with psychological issues such as poor self-image and social . Thus, these effects may contribute to long-term health consequences for individuals even if inhalant abuse ceases, and further research of these consequences relative to inhalant abuse would be beneficial. The adrenal insufficiency hypothesis requires testing in a clinical setting, but if confirmed, presents a significant health risk to those with a history of inhalant abuse, as the disorder can be life-threatening when undiagnosed and untreated. However, this may provide a target for future clinical research, which may improve the health outcomes of highly vulnerable individuals. Collectively, this thesis has utilised multiple methods both clinical and pre-clinical, in order to better understand the health consequences of adolescent inhalant abuse, and to provide translational outputs that will directly improve the detection and treatment of inhalant abuse in adolescents.

2 Rose Crossin (737900)

Declaration

I, Rose Crossin, declare that:

- This thesis comprises my original work towards the Doctor of Philosophy - Due acknowledgment has been made in the text to all other material used - The thesis is fewer than the maximum word limit in length, as approved by the Research Higher Degree Committee

Signed,

Rose Crossin

13 October 2018

3 Rose Crossin (737900)

Preface

Publication status of chapters References for Chapter 1, the Supplementary Section of Chapter 4, and Chapter 5 are reported in the Bibliography. All other references are contained within the Articles tabulated below.

Chapter Article title Status 1 General introduction and literature review Unpublished material not submitted for publication 2 Adolescent inhalant abuse leads to other drug Published by Australian and use and impaired growth; implications for New Zealand Journal of Public diagnosis Health on 23 October 2016 2 The persistence of growth impairments Published by Addiction associated with adolescent inhalant abuse Research and Theory on 15 following sustained abstinence June 2017 3 Growth changes after inhalant abuse and Published by Human and toluene exposure: a systematic review and Experimental Toxicology on 31 meta-analysis of human and animal studies July 2018 4 Adolescent inhalant abuse results in adrenal Accepted for publication in dysfunction and hypermetabolic phenotype Neuroendocrinology on 13 with persistent growth impairments September 2018 5 General discussion Unpublished material not submitted for publication App. 1 Altered body weight associated with substance Published by Addiction abuse: a look beyond food intake Research and Theory on 20 March 2018 App. 2 A proposed clinical cohort exploring the growth Unpublished material not and energy balance effects of adolescent submitted for publication inhalant abuse

Contribution of persons in multi-authored publications

Chapter 2 Sheree Cairney: assistance with data analysis, interpretation, and manuscript preparation.

Andrew Lawrence: assistance with data interpretation and manuscript preparation.

Jhodie Duncan: assistance with data analysis, interpretation, and manuscript preparation.

Chapter 3 Andrew Lawrence: assistance with data interpretation and manuscript preparation.

Zane Andrews: assistance with data interpretation and manuscript preparation.

Leonid Churilov: assistance with meta-analytic techniques, data interpretation and manuscript preparation.

Jhodie Duncan: assistance with data interpretation and manuscript preparation.

4 Rose Crossin (737900)

Chapter 4 Zane Andrews: assistance with experimental design, data interpretation, and manuscript preparation.

Natalie Sims: assistance with skeletal micro-computed tomography and data interpretation.

Terence Pang: assistance with adrenal histology and data interpretation.

Michael Mathai: assistance with faecal bomb calorimetry.

Jonathan Gooi: assistance with bone mechanical testing.

Aneta Stefanidis: assistance with metabolic cage experiments.

Brian Oldfield: assistance with metabolic cage experiments.

Andrew Lawrence: assistance with experimental design, data interpretation, and manuscript preparation.

Jhodie Duncan: assistance with experimental design, data interpretation, and manuscript preparation.

Appendix 1 Andrew Lawrence: assistance with manuscript preparation.

Zane Andrews: assistance with manuscript preparation.

Jhodie Duncan: assistance with manuscript preparation.

Acknowledgement of funding sources The research was supported by the Australian National Health and Medical Research Council (NHMRC) (940835), the Australian Department of Education and Training, from whom Rose Crossin received a scholarship through the Research Training Program, and the Victorian Government’s Operational Infrastructure Support Scheme.

5 Rose Crossin (737900)

Acknowledgements I acknowledge with gratitude my supervisors; Jhodie Duncan, Andy Lawrence and Zane Andrews for their wisdom, support, tolerance and for giving me the freedom to let me develop this project myself. I especially need to acknowledge Jhodie, who has had a couple of tough years herself, but has been an amazing supervisor, mentor, and now I hope I can say, friend. I am also grateful to Julie Bernhardt and Song Yao from my advisory committee for their advice and encouragement.

Many thanks to the addiction lab group at the Florey, who have helped with experiments, provided feedback, and made being here fun. Especially I thank Robyn Brown, Christina Perry, Nikki Chen, and Luba Lee-Kardashyn for their practical advice and support during the tough times.

I acknowledge the contribution of the communities in Arnhem Land, who participated in the inhalant abuse epidemiology studies and who made such positive change to their communities. I also acknowledge Dr Chris Burns, who undertook the majority of the initial data collection, as part of this own thesis.

I worked with an amazing group of collaborators in the last three years and I thank them all for their help and the inspiration that it gave me to work with scientists of their calibre. I also thank the Howard Florey Laboratories animal house staff and Florey histology service for their assistance during my animal experiments.

I acknowledge with love and gratitude my family and friends, who have been there for me during the last three years. Thanks Mum and Dad for teaching me from a young age that things of value do not come easily. Thanks also to my grandparents (Fred and Ivy, Bob and Heather), who wanted to see all of their grandchildren succeed and take up opportunities that they did not have themselves. Especially, thank you with all my heart to my husband Enda, whose practical and emotional support made this endeavour possible. I could not have done this without you.

Lastly, and most importantly, I thank my daughters Zara and Isla, for being the inspiration that kept me here. When I started my PhD they were 1 and 3, and there were plenty of people who tried to discourage me from this path. But I wanted to prove to my girls that family life and careers can co-exist, and so, my darlings, I want you to remember this. When people tell you that you shouldn’t take on great challenges, don’t get angry, don’t get despondent, don’t get offended, get working.

“Energy rightly applied and directed will accomplish anything” – Nellie Bly 1889

(journalist, explorer, badass feminist)

6 Rose Crossin (737900)

This thesis is dedicated to my daughters

Zara and Isla

Thank you for inspiring me and I hope that I can inspire you to follow your dreams and pursue greatness, with hard work, passion, and the support of all those who love you

(Dad and I have got your backs, always and forever)

7 Rose Crossin (737900)

Contents Abstract ...... 1 Declaration ...... 3 Preface ...... 4 Publication status of chapters ...... 4 Contribution of persons in multi-authored publications ...... 4 Chapter 2 ...... 4 Chapter 3 ...... 4 Chapter 4 ...... 5 Appendix 1...... 5 Acknowledgement of funding sources ...... 5 Acknowledgements ...... 6 List of tables, figures, and illustrations ...... 10 Chapter 1 - General Introduction and Literature Review ...... 11 Inhalant abuse ...... 11 Adolescent inhalant abuse ...... 12 The effects of inhalant abuse on food intake, body weight, and growth ...... 13 Energy balance ...... 14 Central and peripheral regulation of energy balance ...... 15 Effects of inhalants on energy intake ...... 18 Effects of inhalants on energy expenditure ...... 18 Effects of inhalants on adipose tissue ...... 19 Effects of inhalants on bone...... 20 Effects of inhalants on the HPA axis and adrenal gland ...... 20 Effects in abstinence and the potential for adult-onset disorders ...... 22 Issues when studying inhalant abuse ...... 22 Summary ...... 23 Research aims and hypothesis ...... 24 Chapter 2 – Growth changes arising from adolescent inhalant abuse in a human cohort ...... 25 Chapter 3 – A systematic review and meta-analysis on the growth impacts of inhalant abuse 39 Chapter 4 – The energy balance consequences of adolescent inhalant abuse and potential causal mechanisms ...... 71 Supplementary Data for Chapter 4 ...... 120 Prelude ...... 120

8 Rose Crossin (737900)

Methods ...... 121 Results ...... 122 Discussion ...... 125 Chapter 5 - General Discussion ...... 127 Adolescent inhalant abuse results in growth suppression and energy balance alterations . 127 The growth impairments arising from adolescent inhalant abuse persist into sustained abstinence ...... 130 Growth changes arising from adolescent inhalant abuse are not attributable to under- nutrition but may be caused by adrenal insufficiency ...... 131 Translational applications of this research ...... 134 Future work ...... 134 Summary and conclusions ...... 136 Bibliography ...... 138 Appendix 1 – Review on the effects of on body weight ...... 147 Appendix 2 – A human cohort of adolescent inhalant users ...... 157 Introduction ...... 157 Methods ...... 157 Current status ...... 160 Preliminary findings ...... 160 Appendix 3 – Research Protocol for the human cohort study ...... 162 Appendix 4 – Questionnaire for the human cohort study ...... 183

9 Rose Crossin (737900)

List of tables, figures, and illustrations Note: this list only includes the figures that are not already published in manuscripts, to avoid duplicated figure numbering. As such, this list includes figures contained within Chapter 1 (general introduction) and supplementary section of Chapter 4.

Figure / Title Section Table number Figure 1 Past year inhalant use in Australian secondary school Chapter 1 students, by gender, based on data sourced from the 2014 Australian Secondary School Drug Survey Figure 2 The energy balance equation Chapter 1 Table 1 Central and peripheral regulation of energy balance Chapter 1 Figure 3 Blood glucose homeostasis Chapter 1 Figure 4 Diagnosis of adrenal insufficiency Chapter 1 Figure 5 Whole body thermographic imaging Chapter 4 supplement Figure 6 IL-1β in the liver and hypothalamus Chapter 4 supplement Figure 7 Energy balance following corticosterone replacement Chapter 4 supplement

10 Rose Crossin (737900)

Chapter 1 - General Introduction and Literature Review

Inhalant abuse Substance abuse or substance use disorders are defined as “chronic, relapsing brain diseases that are characterised by compulsive drug-seeking and use, despite harmful consequences” [1]. Inhalant abuse is a form of substance abuse and involves the intentional inhalation of vapours from household or industrial products, in order to create a feeling of euphoria and an altered mental state [2]. Products that are commonly abused as inhalants include petrol, spray paint, thinners, aerosols, and glue. These products are cheap, legal to purchase, and readily accessible, which is thought to add to their attractiveness as substances of abuse [3].

While inhaled products are not technically considered ‘drugs’, they share the reinforcing nature of many drugs of abuse, due to the presence of toluene, which is common to all these products. Toluene is a volatile solvent; a colourless, water-insoluble liquid that is classed as an aromatic hydrocarbon. Toluene is considered to underlie the addictive drive to continue abusing inhalants, based on its ability to generate a conditioned place preference in rats [4, 5], and preferential inhalation by humans of pure toluene compared with mixed products containing toluene [6]. Therefore, toluene is commonly used as an experimental pre-clinical model for inhalant abuse, although it should be noted that inhaled products contain several harmful compounds [7-9]. Thus, for the purpose of this thesis, the terms “inhalants” and “toluene” are used interchangeably, depending on whether reference is being made to clinical or pre-clinical studies.

Inhalant abuse is commonly known as “chroming”, “huffing”, or “bagging” as common self- administration methods are to inhale concentrated vapours from spray cans, off solvent- soaked cloth, or directly from a bag, respectively [10]. When administered in these ways, the concentration of inhaled toluene is estimated to range from 3,000-15,000 parts per million (ppm) [11]. Once inhaled, the acute effects of toluene occur within minutes; characterised by locomotor stimulation and feelings of euphoria and exhilaration [12, 13]. At concentrations greater than 3,000 ppm, central nervous system (CNS) depression occurs, with acute symptoms including slurred speech, disorientation, weakness, and finally sedation [12, 14]. At its most severe, and usually observed when concentrations exceed 10,000 ppm, CNS depression may progress into cardiopulmonary failure, coma or death [15]. After inhalation, toluene is absorbed across the lungs into the bloodstream before being rapidly metabolised and excreted as urinary hippuric acid [16, 17]. The rapid metabolism of toluene out of the bloodstream makes it particularly difficult to detect, and there are no established blood, urinary or saliva tests for diagnosing or identifying either acute inhalant intoxication or chronic inhalant abuse. While the abuse of inhalants generally occurs for a short duration, ranging from minutes to one hour, for the majority of individuals this behaviour is repeated over time and extends for periods greater than one year, such that exposure becomes chronic but intermittent in nature [18].

The effects of inhalants on the CNS, including cognition, have been relatively well studied [19- 21] and do not form the basis of this thesis. However, inhalants can also have a wide range of peripheral effects throughout the body, including on the renal, cardiovascular, and pulmonary systems [12]. With repeated exposure, toluene accumulates in lipid-rich organs such as the

11 Rose Crossin (737900) brain, adipose tissue, kidneys and adrenal glands [22], and as such, is likely to affect the function of these organs. Additionally, one of the most profound, but under-explored, consequences of inhalant abuse is its effect on food intake, body weight, and growth; especially if exposure occurs during adolescence, suggesting inhalant abuse results in energy balance dysfunction (discussed further beginning page 13) [23-27]. However, understanding of the effects of inhalants on peripheral organs and processes, including energy balance, remains limited. Furthermore, most studies have investigated the acute effects of inhalants at, or shortly following, exposure but have neglected to investigate the long-term effects, even if abuse of inhaled products ceases. Thus, understanding the effects of inhalant abuse on energy balance and growth, including the persistence of these effects in abstinence, form the overarching aim of this thesis. The following review will highlight the current understanding of the effects of inhalants on growth and energy balance.

It is also important to note that, as well as intentional inhalation, exposure to inhaled toluene can also occur in industrial settings (e.g. in factories where toluene is used as a solvent). Both the patterns of use and concentrations of inhaled toluene differentiate inhalant abuse from occupational exposure, with occupational exposure characterised by sustained contact to concentrations of toluene less than 200 ppm [28, 29]. While this thesis focusses on inhalant abuse, it is acknowledged that some of the data pertaining to the toxicology of toluene has been determined using occupational exposure studies and this should be interpreted with caution relative to the effects in the abuse setting; these studies are highlighted when discussed.

Adolescent inhalant abuse Adolescence is defined as the period between puberty and adulthood, typically between the ages of 11 and 21 years [30]. Inhalant abuse is predominantly an issue in young adolescent populations (Figure 1), with prevalence rates approximately equal between males and females [27]. Inhalants are one of the first and most common substances abused by the 12-13 age group in Australia, with 14% of 12 year-olds having misused inhalants in the previous year, and of those, approximately 20% have misused inhalants 10 or more times in that year [27, 31]. Furthermore, this pattern of early adolescent use is consistent globally [32]. Of further concern, a recent survey by the Australian government found that, overall, inhalant abuse in Australia is increasing with rates of recent inhalant use more than doubling between 2007- 2016 compared to a 4% reduction in recent and cigarette use over the same period [31]. Statistics such as these highlight the growing popularity of inhalants as a form of substance abuse and are likely to reflect the increasing rates of inhalant abuse in young adolescents worldwide. For example the most recent US Monitoring the Future Survey also reported a significant increase in recent use in Grade 8 students (aged 13-14) along with a decline in the perceived risk of inhalant use [33].

12 Rose Crossin (737900)

Past year inhalant use in 2014

20

15

10 Male

5 Female % used in past year past in % used 0 12 13 14 15 16 17 Age

Figure 1 – Past year inhalant use in Australian secondary school students, by gender, based on data sourced from the 2014 Australian Secondary School Drug Survey. The data highlight that, amongst all adolescents, the predominant age group misusing inhalants is young adolescents, with use decreasing as age increases. Figure adapted from data provided in [27].

As well as an association with adolescence, inhalant abuse is further associated with vulnerable and disadvantaged groups [34], including indigenous populations. Inhalant abuse (particularly petrol sniffing) remains a significant public health issue for Indigenous Australians [35], with abuse rates of up to 60% reported in some remote communities [36]. Furthermore inhalant abusers are disproportionately represented in the mental health, juvenile justice, and protective services systems [3]. Therefore, given that much of the epidemiological data on inhalant abuse comes from secondary school surveys, it is likely that inhalant abuse rates are actually under-reported in Australia and worldwide. A history of adolescent inhalant abuse also has an association with a range of other behavioural and psychological harms. Adolescents who misuse inhalants are more likely to develop other issues [37, 38], experience major depression [38], and to experience suicidal ideation or attempt suicide [38, 39]. It is important to note, however, that the directionality of these relationships is not known; these may be outcomes of inhalant abuse or arise from the common background of vulnerability and social disadvantage in general.

The effects of inhalant abuse on food intake, body weight, and growth In humans, adolescent inhalant abuse is associated with disordered eating [26], weight loss [24], and emaciation [40]. Inhalant abuse in both males and females has been associated with fasting, purging, and other behaviours associated with attempts to lose weight [26], with previous studies suggesting that inhalants may be used as a means of suppressing appetite and thus body weight [26, 41]. These findings are consistent with pre-clinical studies using rats, with adolescent and adult toluene exposure (repeated exposures, including intraperitoneal and inhalation administration, concentrations ranging from 3,000-16,000 ppm) associated with reduced food intake, decreased weight gain, and reduced fat deposition [25, 42, 43]. Indeed, the effect of inhalant abuse on body weight is so well known that it is listed as a warning sign of inhalant abuse [44].

13 Rose Crossin (737900)

In addition to effects on food intake and body weight, exposure to toluene is associated with impaired skeletal growth. In weanling rats, chronic toluene exposure at concentrations analogous to inhalant abuse (6,200 ppm, 15 mins/hour, 8 h/day, for 23 weeks) has been shown to impair skeletal growth, with a significant reduction in rump width and variable reduction in torso length [9]. Skeletal variations have also been shown in the offspring of rats where there was maternal exposure to toluene (up to 2,000 ppm, 6 h/day, 7 d/week for 80 days premating and 15 days of mating), as well as decreased body weight in the offspring [45, 46]. In humans, regular inhalant abuse (spray paint inhalation) during pregnancy is also associated with neonatal skeletal malformations and growth impairment [47].

As well as a reduction in overall skeletal growth, inhalant abuse is also associated with reduced bone density. A study of adolescent males with a history of chronic glue sniffing (mean duration of abuse 3.2 years) found that bone mineral density was significantly reduced in comparison to an age-matched non-sniffing group [48]. In this study, the mean height of the glue sniffing group was reduced in comparison to controls, though this result was not significant [48]. However, no other human studies on the effects of adolescent inhalant abuse on skeletal growth could be found. Collectively, these studies suggest that toluene exposure can lead to reductions in food intake, body weight suppression, and impaired growth. However, both the mechanisms underlying these consequences of adolescent inhalant abuse, and the persistence and long-term health effects of these changes in abstinence are currently unknown.

The impact of inhalants on food intake, body weight, and growth is of particular importance given the association between inhalant abuse and adolescence. During adolescence, critical maturational processes occur, including an intensive period of growth known as the adolescent growth spurt [49], where skeletal structures grow and mature [50]. Indeed, in humans, over half of the peak bone mass is attained during the adolescent growth spurt [50, 51]. However, the adolescent growth spurt can be disrupted by factors such as malnutrition [52] or exposure to estrogenic chemicals [53]. Growth impairments can potentially recover if the harmful factor is removed, but if exposure occurs during adolescence usually only partial recovery is possible [54]. Thus, any disruption to growth during adolescence has the potential to cause long-term effects on growth patterns [55]. Given that the highest rates of inhalant abuse are occurring during a critical maturational period, it highlights the importance of studying the impacts of inhalant abuse specifically in adolescents, including the long-term effects even if exposure ceases.

Energy balance Food intake, body weight, and growth are intrinsically linked and can be represented via the energy balance equation (Figure 2), with body weight a function of the energy stored in the body. Thus, an imbalance in this equation can lead to changes in body weight. Energy intake is primarily driven by food intake and moderated by absorption and excretion. Energy expenditure includes basal metabolic rate, physical activity and thermogenesis (subdivided into cold-induced thermogenesis and the thermic effect of food). Energy balance is controlled by the processes of metabolism, which encompasses a system that regulates eating behaviour and nutrient absorption, as well as the storage and release of energy – termed nutrient partitioning. Body weight will remain stable when there is a balance between energy intake,

14 Rose Crossin (737900) storage, and expenditure, known as energy homeostasis. Energy homeostasis is the balance between the pathways that stimulate food intake and promote weight gain, and the pathways that reduce food intake and promote weight loss [56]. The maintenance of body weight is buffered, i.e. a decrease in energy intake results in decreased expenditure and vice versa, therefore, any changes in body weight occur by overwhelming the buffering potential of homeostatic systems [57].

Figure 2 – The energy balance equation. Original figure by R Crossin.

A negative energy balance occurs when energy intake is less than energy expended, and this shortfall is made up by a release of energy from body stores (e.g. adipose tissue), leading to reduced body weight. If the difference between energy intake and energy expenditure is positive, then energy is stored and body weight increases (positive energy balance). Physiological processes strongly defend reductions in body weight arising from a negative energy balance, ultimately leading to weight regain [58]. In contrast, a positive energy balance that leads to weight gain does not initiate the same physiological defence, or resistance to that physiological defence develops, and weight gain can continue into obesity. This new increased body weight is subsequently defended [59]. Energy balance becomes particularly important in periods of rapid growth, such as the adolescent growth spurt, with energy intake one of the major determinants of growth [60, 61]. However, the effect of energy intake on growth is predominantly indirect, with growth impairments due to under-nutrition primarily attributable to endocrine changes such as increased circulating cortisol [62-64].

Central and peripheral regulation of energy balance Energy homeostasis, and thus body weight, is both centrally and peripherally regulated through the processes of metabolism [56], as described in Table 1. In order to understand the potential mechanisms behind inhalant-induced energy balance dysfunction, it is important to summarise the complex neuroendocrine system that regulates energy balance. The main short term fuel source of the body is glucose, which is maintained within highly-regulated levels [65]. Excess glucose in the blood is stored in the liver and muscles as glycogen, in response to insulin release, via the process of glycogenesis. Glycogen is then available for release in response to falling blood glucose levels, via the process of glycogenolysis, which is stimulated by the Hypothalamic-Pituitary-Adrenal (HPA) axis. Hypoglycaemia (low blood glucose) is a life- threatening condition, and thus the response is rapid and involves both central and peripheral regulation [65]. This counter-regulatory response activates both feeding behaviour and the release of glycogen stores, in order to rapidly normalise blood glucose levels [66], summarised in Figure 3.

15 Rose Crossin (737900)

Table 1 – central and peripheral regulation of energy balance

Key metabolic organ Compound Function Pancreas Insulin Decreases glucose levels in the blood and promotes lipogenesis (the storage of energy as adipose tissue) [67] Pancreas Amylin Slows gastric emptying and reduces appetite [68] Liver Insulin Responds to insulin secreted by the pancreas by taking up glucose from the blood [69] Kidney Insulin Clears and degrades circulating insulin [70] Gut Ghrelin Increases hunger and initiates feeding [71] Gut Mechanical Stretch receptors signal satiety to the brain via processes the vagus nerve [72] Gut Peptide YY (PYY) Slows gastric emptying and reduces appetite [73] Adipose tissue Leptin Opposes the actions of ghrelin, by sending an adiposity signal and inhibiting hunger [74] and also inhibits lipogenesis [75] Brain Receptors for gut Signals converge primarily on the brain stem hormones and hypothalamus, which are the primary brain regions involved in maintaining energy homeostasis [76] Brain Neuropeptide Y Increases food intake [77] (NPY) Adrenal gland Adrenalin and Responds to hypoglycaemia by inhibiting cortisol insulin release, and activates glycogenolysis and gluconeogenesis [65] Bone Osteocalcin Increases insulin production and sensitivity, and enhances glucose utilisation [78]

Insulin release also promotes the conversion of excess glucose to fatty acids, via the process of lipogenesis, which is then stored in adipose tissue [79]. Adipose tissue is thus the major site of long-term energy storage, noting however that some fatty acids can be stored ectopically in organs such as the liver. When energy homeostasis is achieved, the amount of energy stored as adipose tissue is stable [76], but during a negative energy balance, energy can be released by the breakdown of adipose stores via the process of lipolysis. The lipostatic model of energy balance states that food intake is not tied to the immediate energy needs, but rather it is regulated by signals that are proportional to the size of the body’s adipose stores [80]. Thus, weight loss caused by the depletion of energy stored as adipose tissue should increase food consumption. According to the lipostatic model, in response to weight loss that arises from insufficient energy intake, levels of the regulatory hormones insulin and leptin will decrease, which activates pathways that stimulate appetite and promote weight gain [56].

16 Rose Crossin (737900)

Figure 3 – Blood glucose homeostasis. Maintenance of blood glucose levels, whereby excess blood glucose is stored in tissue, and released via counter-regulatory responses to hypoglycaemia, image adapted from [81]

When metabolic processes are functioning correctly, feedback mechanisms correct disruptions to energy homeostasis, with both the hypothalamus and brain stem being key regions involved in sensing both short and long term changes in energy balance. The response to a negative energy balance is firstly the promotion of appetite / feeding behaviour, and the utilisation of glycogen stores (Figure 3), and when these are exhausted, the utilisation of adipose tissue stores, and finally the breakdown of lean muscle mass occurs. However, as described, adolescent inhalant abuse is associated with both decreased food intake, impaired weight gain, and fasting hypoglycaemia [23], which suggests that energy homeostasis is not being achieved and the feedback mechanisms, which should correct a negative energy balance, are dysfunctional.

Toluene has a complex pathway through the body. As previously discussed, once inhaled, toluene crosses rapidly from the lungs into the bloodstream, before being metabolised by the liver and excreted by the kidneys [16, 17]. With repeated exposure, toluene can accumulate in organs, primarily the brain, adipose tissue, and adrenal glands [22, 82]. As shown in Table 1, these are key regions for metabolic processes. Toluene can also cross [83] and disrupt [84] the blood-brain barrier and has a comparatively high accumulation in the brainstem and hypothalamus [85]. The brainstem and hypothalamus play a critical role in control of feeding and metabolism [76, 86], which raises the potential that inhalant-induced metabolic dysfunction may have both centrally and peripherally mediated components.

17 Rose Crossin (737900)

The symptoms of inhalant abuse (i.e. reduced food intake, body weight suppression, and impaired growth) are suggestive of a negative energy balance. However, it is not clear which components of the energy balance equation are altered, or how the central and peripheral regulation of energy balance may be being affected. Theoretically, inhalant abuse could reduce energy intake via a direct effect on appetite, or by increasing energy expenditure via changes to physical activity, basal metabolic rate, or thermogenesis. Inhalant abuse may also disrupt the metabolic signalling that would promote appetite and weight gain in response to a negative energy balance. Alternatively, exposure to inhalants could also be directly affecting body composition through effects on the absorption or deposition of fat, or skeletal composition, which may explain the observed growth impairments [9]. Each of these aspects of energy balance will be expanded upon below, as they are key components to explore in order to understand the energy balance and growth effects of inhalant abuse.

Effects of inhalants on energy intake In humans there is evidence that incidental inhalation of petrol fumes may have an acute anorectic effect, and the study’s authors hypothesised that in a food-constrained environment, a desire to suppress feelings of hunger could be a driver for intentional petrol sniffing behaviour [87]. These data confirmed previous anecdotal reports that petrol sniffing in Australian Indigenous communities is partly driven by a desire to suppress appetite [41], although the relationship between appetite suppression and inhalant abuse among street children in India is less consistent [88]. In adult rats, toluene-induced anorexia (intraperitoneal administration, 1.3 mL/kg/day, for 4 days) has been attributed in part to the observed decrease in the hypothalamic expression of Neuropeptide Y (NPY), a peptide which stimulates food intake [25]. Furthermore, energy intake is moderated by absorption of nutrients occurring in the gut, and disruptions to absorption are known to be associated with other types of substance abuse [89]; however, nutrient absorption across the gut following toluene exposure or inhalant abuse has never been assessed. This is a potentially significant knowledge gap, given that inhalant users frequently report gastrointestinal disturbances, including nausea, vomiting and diarrhoea [12, 90] and the potential effects on the gut microbiome have never been assessed in relation to inhalant abuse.

There is also evidence to suggest that inhalant abuse can impair metabolic signalling, because the normal responses to negative energy balance do not occur. Our lab has shown that food intake is reduced in rodents following exposure to toluene at abuse concentrations, with animals exposed to toluene consuming fewer kilo-calories than air-exposed animals at the same body weight [23]. This finding suggests that there is impairment to the metabolic feedback processes, which should act to restore homeostasis in response to insufficient energy intake. In rats, adolescent exposure to toluene (10,000 ppm, 1 h/day, 3 d/week, for 4 weeks) has been shown to alter levels of gut and pancreatic hormones, including insulin, amylin, and Peptide YY (PYY), as well as decrease hypothalamic leptin receptor mRNA expression [23]. Importantly, decreased amylin and PYY levels should act to increase appetite and food intake, but this response does not occur, supporting the idea of impaired metabolic signalling.

Effects of inhalants on energy expenditure Energy expenditure in an animal model of inhalant abuse has never been explicitly tested, but studies suggest that exposure to toluene affects components of energy expenditure (i.e.

18 Rose Crossin (737900) physical activity, basal metabolic rate, and thermogenesis). Toluene exposure in adolescents acutely increases locomotor activity (exposure by inhalation at 4,000 ppm) [7], although this effect is transient and the response to toluene is bi-phasic and locomotor activity is reduced at higher concentrations [91]. In relation to basal metabolic rate, a human study of occupational exposure to toluene found that chronic exposure (less than 100 ppm) was associated with increased food intake, though this was not reflected in an increased body mass index (BMI) [92]. The authors hypothesised that toluene exposure resulted in a higher basal metabolic rate, though this was not tested [92]. In contrast, animal studies (pregnant female rats, exposed to toluene by inhalation at 8,000, 12,000 and 16,000 ppm twice daily for 14 days) showed that pre-natal exposure to toluene resulted in reductions in metabolic rate and energy expenditure in the offspring, unfortunately this was not also assessed in the toluene-exposed individuals [42]. These studies also did not assess thermogenesis, in addition to physical activity or metabolic rate, but one study utilising mice (inhaled toluene at 2,000, 4,000 and 6,000 ppm once for 60 mins) found that toluene acutely reduced core temperature in a dose dependent manner [93]. Conversely, it is interesting to note that many inhalant users report a persistent low-grade fever [94] though a mechanism for this remains unknown.

Effects of inhalants on adipose tissue Adipose tissue is the major site of long-term energy storage in the body, and changes to adiposity will be reflected in changes to body weight. Therefore, adipose tissue is a key organ to consider in relation to adolescent inhalant abuse. In our laboratory’s rodent model of adolescent inhalant abuse (10,000 ppm, 1 h/day, 3 d/week, for 4 weeks), body composition was altered by toluene exposure. Male rats exposed to toluene had significantly reduced adiposity but no difference in lean mass or total water [23]. Interestingly, in offspring that had prenatal exposure to toluene (0-16,000 ppm, 15 mins twice/day, gestational days 8-20), body weight was also reduced, but toluene significantly increased adiposity in a dose-dependent way in male, but not female, offspring [42]. The increased adiposity was attributed to a compensatory developmental programming effect, whereby low birth weight alters hormonal and metabolic programming to maximise postnatal survival, and subsequently results in increased adiposity [95]. Furthermore, when there is a negative energy balance caused by insufficient food intake, adipose stores are utilised by the body to provide energy. Thus, the observed decrease in adiposity [23] associated with exposure to toluene may not be unexpected. However, alternative hypotheses as to why toluene reduces adipose levels are that it affects the absorption of ingested fats within the digestive tract, or the ability to depose lipids in adipose tissue, such as that observed in pancreatic disorders [96]. For example, both leptin and insulin play a crucial role in regulating lipogenesis [79] and therefore the observed disruptions to hormones involved in energy balance signalling, including insulin [23], may impair the deposition of lipids in adipose tissue. While the studies discussed above show that exposure to toluene has an effect on body composition, particularly adiposity [23], it has yet to be determined whether toluene has a direct effect on body composition, or if these changes are an indirect effect arising as a result of decreased food intake leading to a negative energy balance.

19 Rose Crossin (737900)

Effects of inhalants on bone The reduction in weight gain arising from toluene exposure may not only be due to reduced adiposity, but also due to skeletal growth impairments. Human studies investigating the effects of toluene on skeletal growth are extremely limited and therefore no firm conclusion can be currently reached on the acute and chronic impacts of adolescent inhalant abuse. However, is also associated with impaired bone function and subsequent increased osteoporosis and fracture risk [97, 98]. Given that alcohol has a known metabolic effect [99] and similar modes of action to toluene [100, 101], this would suggest that the skeletal impacts of inhalant abuse warrant further study. Anorexia nervosa (AN) can provide another biologically relevant model for skeletal function, with symptoms of reduced food intake and fat deposition, occurring predominantly in adolescence [102], as has been observed in inhalant abusers. Impaired skeletal growth and function is a common symptom of AN [103], which has been associated with low leptin levels [104]; a common feature of AN [105]. Of major concern, the loss of bone density that is observed in adolescents with a history of AN does not resolve, even years after weight restoration [106, 107].

There are a number of potential mechanisms that may lead to impaired skeletal growth arising from adolescent inhalant abuse. Male adolescent mice exposed to toluene (300 ppm, 6 h/day, for 8 weeks) have been shown to have significantly reduced bone mineral density compared to controls [108]. As bone metabolism occurs as a continuous cycle, whereby osteoclasts resorb existing bone and osteoblasts then lay down replacement bone, the authors of this paper hypothesised that toluene may be disrupting both of these processes [108]. The metabolic perturbations that are observed in inhalant abuse could also be involved in the reported skeletal growth impairments. Leptin has a direct effect on bone cell function and acts to reduce bone fragility through anabolic effects on osteoblasts and the reduction of osteoclast formation [109]. Amylin increases bone mass by inhibiting bone resorption [110]. Insulin has an indirect effect on bone function, and increased circulating insulin is associated with increased bone density [111]. All of these hormones are affected following exposure to toluene [23, 112].

Bone itself can also exert reciprocal effects upon metabolism via osteocalcin, which increases insulin production and sensitivity, and enhances glucose utilisation [78, 113]. Osteocalcin can exert effects on both pancreatic β cells and adipocytes, making it a key component of metabolic regulation [114]. In combination, these developments led to the conclusion that there is reciprocal regulation between energy metabolism and bone metabolism [115-117]. Furthermore, this is mediated via the hypothalamus [118] which is known to be affected by exposure to toluene [85]. Given the metabolic functions of bone, any investigation into the metabolic disruptions caused by inhalant abuse should therefore also give consideration to the skeletal system. Furthermore, data from AN models suggest that if inhalant abuse does have a skeletal effect, it will be important to study the chronic impacts, as it is likely that the effects will persist in abstinence.

Effects of inhalants on the HPA axis and adrenal gland There is evidence to suggest that inhalant abuse may have an effect on the HPA axis, though the nature of this relationship is equivocal. Some studies have associated inhalant abuse with an up-regulation in HPA axis activity and associated behaviours [119-121], whereas others

20 Rose Crossin (737900) have found a blunted HPA response following inhalant abuse [122, 123]. It is also possible that there is a direct effect of toluene on the adrenal gland, which is where the hormones cortisol and adrenaline are produced as part of the stress response, in the zona fasiculata of the cortex and the medulla, respectively. The adrenal gland is vulnerable to toxicological injury due to high vascularity and fat content [124], particularly by aromatic hydrocarbons such as toluene [125]. Multiple pre-clinical studies have found that adrenal hypertrophy is evident following inhalant abuse [23, 126, 127], though only one clinical case study has associated inhalant abuse with an adrenal effect [128].

Adrenal hypertrophy can be an outcome of chronic stress or of adrenal insufficiency [129]. Adrenal insufficiency is a failure to produce the stress hormone cortisol by the cortex of the adrenal gland, either due to an issue in the adrenal gland (primary adrenal insufficiency) or impairment upstream in the HPA axis (secondary or tertiary adrenal insufficiency) [130]. Chronic stress is characterised by adrenal hypertrophy with elevated levels of cortisol, whereas adrenal insufficiency is characterised by adrenal hypertrophy with low or unchanged levels of cortisol, which is then further located within the HPA axis by the relative levels of the other HPA axis signalling hormones adrenocorticotropic hormone (ACTH) and corticotropic-releasing hormone (CRH) [129, 130], as shown in Figure 4.

Figure 4 – Diagnosis of adrenal insufficiency. The differentiation between chronic stress and adrenal insufficiency, and the localisation of adrenal insufficiency within the HPA axis by the relative quantities of cortisol (Cort), adrenocorticotropic hormone (ACTH) and corticotropic-releasing hormone (CRH), and responses to an insulin tolerance test (ITT). Original figured by R Crossin adapted from data in [129, 130].

21 Rose Crossin (737900)

When the adrenal gland has been directly investigated in relation to inhalant abuse, studies have found adrenal hypertrophy, particularly in the zona fasiculata of the adrenal cortex where cortisol (corticosterone in rats) is produced, and low/unchanged levels of basal corticosterone with high levels of basal ACTH [126, 127]. It is interesting to note that although these findings are diagnostically consistent with primary adrenal insufficiency [130], in both studies the authors attributed their findings to chronic stress [126, 127]. Adrenal insufficiency is a chronic disorder with symptoms of decreased food intake, impaired body weight and growth, non- diabetic fasting hypoglycaemia, persistent low-grade fever, fatigue, and gastrointestinal disturbances [130]. These symptoms are consistent with what has been observed following inhalant abuse [23, 24, 26, 90, 94, 131], though it is acknowledged that they also fit a range of other disorders. Nevertheless, there is potential that HPA axis dysfunction, particularly adrenal insufficiency, may underlie the growth and energy balance consequences of inhalant abuse.

Effects in abstinence and the potential for adult-onset disorders Pilot data from our laboratory suggest that some of the energy balance effects arising from adolescent inhalant abuse, including reduced body weight, persist into abstinence [23]. Severe and chronic weight suppression is associated with cognitive impairments [132] and health effects, including kidney and bone damage [133]. Additionally, growth impairments are associated with adverse psychological consequences, including and poor self- image [134, 135]. Conversely, weight regain after a period of weight suppression is associated with increased central adiposity and insulin resistance, both of which are risk factors for the development of metabolic syndrome, obesity, and type-2 diabetes whereby tissues become resistant to insulin [58, 136]. It is important to note, however, that it is currently unknown whether weight suppression arising from adolescent inhalant abuse ultimately resolves. Nevertheless, adolescent inhalant abuse has the potential to initiate a sequence of long-term metabolic alterations and thus lead to a chronic disease burden for individuals, even if exposure ceases. However, the nature of that disease burden remains unknown and knowledge on the medical consequences of inhalant abuse in abstinence is scarce.

Current treatments for substance abuse focus primarily on cessation of use, and energy balance impacts are given limited treatment consideration beyond nutritional counselling in abstinence [137, 138]. Energy balance dysfunction is an under-explored aspect of chronic drug use, but inhalant abuse is not the only drug of abuse that has an energy balance impact. , alcohol, methamphetamine, and have all been linked to metabolic dysfunction [99, 139-141]. Furthermore, metabolic symptoms are relatively well reported in relation to substance abuse, including weight loss during active drug use, and weight gain in abstinence (see for review [142] and [143] provided as Appendix 1). Thus, understanding the link between adolescent inhalant abuse and energy balance dysfunction may provide important knowledge on the long-term consequences of both inhalants and other drug use and lead to treatment strategies that extend beyond drug use cessation.

Issues when studying inhalant abuse Understanding of the potential effects of inhalant abuse is constrained by limited data, and the diversity of exposure models and experimental subjects used. When interpreting human data, some caution must be used due to the confounds of other substance abuse [18]. This is particularly relevant as inhalant abuse is associated with an increased frequency of other drug

22 Rose Crossin (737900) use [144]. For example, the metabolism of toluene can be inhibited by the concurrent consumption of alcohol, which adds a confound to the understanding of whole-body effects [145]. Additionally, laboratory studies into the acute effects of inhalant abuse in humans are generally conducted in adults, despite adolescents being the predominant population abusing inhalants. This is important as the response of adolescents to drugs of abuse is not the same as that of adults [146, 147] and this also holds true for inhalant abuse. For example, adolescents have a decreased sensitivity to the locomotor effects of toluene in models of chronic toluene exposure between 2,000 and 16,000 ppm [7, 91] and interestingly, sensitivity continues to increase throughout adulthood, with oldest rats having the greatest sensitivity to the effects of toluene [148]. However, while adolescents are less sensitive to psychomotor effects of toluene, they are more vulnerable to its toxic properties [149]. Additionally, adolescents take longer than adults to recover from the acute intoxicating effects of toluene (single exposure at 5,000 ppm for 15 or 30 mins) [150]. Therefore it is difficult to directly extract data from studies using adult participants and relate the outcomes to adolescent inhalant users. Finally, the majority of human studies on the effects of toluene exposure have been predominantly conducted using low concentrations of toluene. In some cases this is because researchers were investigating occupational exposure to toluene and so the concentrations used were less than 200 ppm [28, 29], however, when deliberately inhaled, concentrations of toluene can be up to 15,000 ppm [11].

Therefore, it is important to specifically study adolescents in relation to inhalant abuse because:

- they are the predominant population abusing inhalants; - they need to inhale higher concentrations to achieve the same effect as adults, due to decreased sensitivity; - they are more vulnerable to the toxic properties of toluene; - they are abusing inhalants during a critical growth and maturational period; and - the long-term effects, even if exposure should cease, have not been explored.

Summary The cognitive and neurological consequences of inhalant abuse have dominated inhalant abuse research. Long-term cognitive impairment arising from intentional inhalation of inhalants containing toluene is well reported in both human [19-21] and animal [8] studies. In extended abstinence, these effects generally improve and in some cases normalise completely [43, 151, 152]. Although the primary research focus of inhalant abuse has historically been the brain, inhalant abuse is also associated with a range of symptoms that occur throughout the body. The inhalation of toluene is associated with gastrointestinal complaints, including nausea and vomiting [90], as well as more widespread issues such as fatigue [131] and persistent low-grade fever [94]. As discussed, inhalant abuse is associated with decreased food intake, reduced body weight, fasting hypoglycaemia, altered levels of gut hormones, as well as a preliminary link to impaired skeletal growth. Collectively, these symptoms suggest widespread energy balance dysfunction, which can impact growth, and may lead to an increased risk of chronic metabolic disorders e.g. obesity and type-2 diabetes in adulthood, even if abuse ceases. These potential energy balance and growth impacts are not well-

23 Rose Crossin (737900) researched, either acutely or in abstinence, and it is unknown what the long-term health consequences of inhalant-induced energy balance dysfunction may be.

Research aims and hypothesis Based on the information presented above, the aims of this research were to:

- characterise the energy balance and growth consequences of adolescent inhalant abuse; - understand how these effects may persist into abstinence; and - elucidate the mechanisms underlying these changes.

The hypothesis was that adolescent inhalant abuse will cause energy balance dysfunction and growth impairments that will persist into abstinence.

This research specifically addressed the following questions:

1. How does adolescent inhalant abuse affect the energy balance equation and metabolic signalling of energy balance? 2. Do the energy balance and growth consequences of adolescent inhalant abuse resolve or persist in abstinence? 3. Are the observed changes solely due to changes in food intake i.e. inhalant-induced under-nutrition, or are there additional mechanisms?

In order to fulfil these research aims, epidemiological data from an Australian Indigenous population who chronically abused inhalants during adolescence, followed by sustained abstinence of up to 15 years [144, 151, 152] was analysed. A systematic review and meta- analysis on the growth effects of inhalant abuse and toluene exposure was conducted, in order to determine the consistency of growth effects across studies and potential moderators of growth effects. The epidemiological data was analysed in conjunction with an established rodent model of adolescent inhalant abuse (10,000 ppm, 1 h/day, 3 d/week, for 4 weeks). This model mimics the chronic and intermittent nature of abuse seen in humans and has resulted in behavioural outcomes similar to those reported in human abusers [8]. The validity of using rats for inhalant studies is further supported by the fact that rats and humans have an equivalent sensitivity to toluene, and thus, the effects of toluene in humans may be estimated using rat models [153].

For the rodent studies, toluene was used in isolation as it is the common volatile solvent in abused products and is preferentially inhaled when available [6]. The combined power of epidemiological data with a well-established rodent model allowed characterisation of the energy balance and growth changes specifically associated with the inhalation of toluene and the mechanisms underlying these changes; providing a unique insight into the long-term consequences of adolescent inhalant abuse. Thus, this project addresses an important knowledge gap about the long-term energy balance and growth impacts of adolescent inhalant abuse, including whether these changes persist even if exposure ceases. Ultimately it is hoped that this information will further elucidate the long-term health risks associated with adolescent inhalant abuse and assist clinicians to treat individuals with a history of inhalant abuse, subsequently improving their quality of life.

24 Rose Crossin (737900)

Chapter 2 – Growth changes arising from adolescent inhalant abuse in a human cohort Chapter 2 of this thesis is based on a retrospective analysis of data collected from a cohort of Indigenous males in Australia, who sniffed petrol during adolescence. The aim of this study was to describe the growth changes arising from inhalant abuse, both during active inhalant abuse and after sustained abstinence. The rationale for this aim was two-fold. Firstly, weight impairments arising from adolescent inhalant abuse were relatively well-described in both clinical and pre-clinical studies, however given the adolescent growth spurt is characterised by dramatic increases to both weight and height, it was reasonable to hypothesise that inhalant abuse may also deleteriously affect height. Yet very few studies had investigated the effects of inhalant abuse on linear growth, with inconsistent findings between clinical and pre-clinical studies. This was described in the first paper: Adolescent inhalant abuse leads to other drug use and impaired growth; implications for diagnosis.

Additionally, data from pre-clinical studies had suggested that the weight impairments from inhalant abuse persisted into sustained abstinence; however, this had not been investigated in a human cohort of inhalant users. This was investigated in a sub-set of the same cohort of inhalant users, who had been abstinence from inhalant abuse for two years. This was described in the second paper: The persistence of growth impairments associated with adolescent inhalant abuse following sustained abstinence.

Paper citations:

Crossin, R., Cairney, S., Lawrence, A. J., & Duncan, J. R. (2017). Adolescent inhalant abuse leads to other drug use and impaired growth; implications for diagnosis. Australian and New Zealand Journal of Public Health, 41(1), 99-104.

Crossin, R., Cairney, S., Lawrence, A. J., & Duncan, J. R. (2018). The persistence of growth impairments associated with adolescent inhalant abuse following sustained abstinence. Addiction Research & Theory, 26(3), 183-186.

25 Adolescent inhalant abuse leads to other drug use and impaired growth; implications for diagnosis

Rose Crossin,1 Sheree Cairney,2,3 Andrew J. Lawrence,1,4 Jhodie R. Duncan1,4

nhalant abuse involves the intentional Abstract inhalation of vapours from products, such as petrol and glue, to create a feeling Objective: Abuse of inhalants containing the volatile solvent toluene is a significant public I 1 of euphoria and an altered mental state. health issue, especially for adolescent and Indigenous communities. Adolescent inhalant These products are cheap, legal and readily abuse can lead to chronic health issues and may initiate a trajectory towards further drug accessible, adding to their attractiveness use. Identification of at-risk individuals is difficult and diagnostic tools are limited primarily to as substances of abuse. A recent stratified measurement of serum toluene. Our objective was to identify the effects of adolescent inhalant and randomised selection survey of more abuse on subsequent drug use and growth parameters, and to test the predictive power of than 26,000 people aged 12 years and older growth parameters as a diagnostic measure for inhalant abuse. from across Australia, by the Australian Methods: We retrospectively analysed drug use and growth data from 118 Indigenous males; government, found a 23% increase in inhalant 86 chronically sniffed petrol as adolescents. abuse,2 highlighting its growing popularity. Results: Petrol sniffing was the earliest drug used (mean 13 years) and increased the likelihood In Australia inhalant abuse, especially petrol and earlier use of other drugs. Petrol sniffing significantly impaired height and weight and was sniffing, remains a significant issue for associated with meeting ‘failure to thrive’ criteria; growth diagnostically out-performed serum Indigenous communities with rates up to toluene. 60% in some remote communities.3 However Conclusions: Adolescent inhalant abuse increases the risk for subsequent and earlier drug in recent years, inhalant abuse has also use. It also impairs growth such that individuals meet ‘failure to thrive’ criteria, representing an become prevalent among young adolescents, improved diagnostic model for inhalant abuse. irrespective of ethnicity, as determined by both the household drug survey and Implications for Public Health: Improved diagnosis of adolescent inhalant abuse may lead to a specialised survey of 25,000 Australian earlier detection and enhanced health outcomes. secondary school students aged 12-17.2,4 This Key words: petrol sniffing, height, weight, Failure to Thrive is concerning as adolescence encompasses a critical period of maturation, which for a diagnosis to be made, an individual metabolised, which limits its diagnostic can be disrupted by exposure to harmful needs to be in the care of a psychological capabilities.8 As creatine kinase is elevated in substances.5 Inhalant abuse during this or health professional. Therefore, the first response to petrol sniffing it has also been period is associated with long-term adverse level of recognition of inhalant abuse often suggested as a potential diagnostic tool, health outcomes affecting both central and relies on a high index of suspicion from though its relevance to inhalants other than peripheral processes, including cognitive parents/guardians, teachers or health care petrol remains unknown.9 For most users, dysfunction, renal tubular acidosis, hepatic workers before referral to an appropriate the pattern of inhalant abuse is chronic but dysfunction and bone marrow suppression.6 health professional can occur, which intermittent,10 which highlights the need for Despite its prevalence and associated makes diagnosis problematic. Additionally, diagnostic markers that are more responsive harms, inhalant abuse remains a poorly inhalant abuse rates are typically highest in to sustained use. Chronic inhalant abuse recognised problem.6 There are reported populations of vulnerable youth, which have is associated with long-term health and warning signs to identify inhalant abuse, but reduced contact with the primary health psychological harm including increased these refer predominantly to acute effects, care system.7 Toluene is the primary volatile use of other drugs,11 cognitive decline and which are highly transient.6 The Diagnostic solvent in abused products, and therefore brain injury12 and adverse psychological and Statistical Manual of Mental Disorders serum toluene can be used as an acute consequences including suicide.13 Inhalant IV contains criteria for inhalant abuse, but diagnostic test. However, toluene is rapidly abuse, especially during adolescence,

1. Behavioural Neuroscience, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Victoria 2. Centre for Remote Health, Flinders University, Northern Territory 3. Ninti One Limited, Northern Territory 4. Department of Anatomy and Neuroscience, University of Melbourne, Victoria Correspondence to: Mrs Rose Crossin, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Vic 3010; e-mail: [email protected] Submitted: March 2016; Revision requested: May 2016; Accepted: June 2016 The authors have stated they have no conflict of interest. Aust NZ J Public Health. 2016; Online; doi: 10.1111/1753-6405.12595

2016 Online Australian and New Zealand Journal of Public Health 1 © 2016 Public Health Association of Australia Crossin et al.

can also affect peripheral body systems, 2274). Data were collected from 1992 to 12 hours of their most recent petrol sniffing including those that regulate metabolism 1994, from 118 Indigenous males from two activity, which was verbally confirmed with and growth.14-16 Thus a more robust remote Indigenous communities in northern participants by the researcher during the diagnostic method, which can be applied Australia. Eighty-six participants had a history interview. Samples for blood hydrocarbons to chronic inhalant abuse, may increase of petrol sniffing as adolescents (both leaded (e.g. toluene) were collected into 7mL sterile, detection and provide an opportunity for and unleaded petrol) and 32 participants EDTA Vacutainers; all other haematological intervention strategies, to limit adverse from the same communities with no history measures collected into sterile (EDTA, heparin neuropsychological, metabolic and growth of petrol sniffing acted as age-matched clotted Vacutainers). All haematological outcomes. controls. Informed written consent was given variables and analysis methods are shown in Profiles of subsequent drug use and growth by all participants, who could withdraw from Supplementary Table 1. impairment arising from adolescent inhalant the study at any time. At the time of data abuse are compelling targets for further collection the average age of participants was Parameters of growth research, given that their incidence may 20.6±0.4 years. Height (cm) and weight (kg) were measured contribute to psychological harm and by the research team at the time of the chronic health effects. This study aimed to Drug use survey interview and body mass index (BMI) characterise patterns of other drug use and Classification into petrol sniffing or control calculated (weight (kg)/height squared (m2)). effects on growth in a cohort of adolescent groups was based upon a consensual Weight was measured on an electronic scale inhalant abusers from a population methodology, requiring corroboration from and height was measured using a tape. of Indigenous males. It also aimed to the individual, community health workers, These measures were undertaken by a single understand whether growth parameters research assistants and local health records. trained individual, to prevent inter-rater could represent a diagnostic tool to identify Each participant’s self-reported history was variability. Growth percentiles were calculated chronic inhalant abuse in adolescents, compared with local community clinic health using US Centre for Disease Control (CDC) including whether the effects were severe records, an assessment by a community 2000 growth charts (males aged 0-20 years19) enough to meet clinical ‘failure to thrive’ (FTT) health worker, and an assessment of petrol and the CDC National Health and Nutrition criteria. sniffing behaviour by research assistants Examination Survey (males > 20 years20). The recruited from the same language group. If criteria for meeting a FTT diagnosis were rd Method data was consistent across all these sources, weight below the 3 percentile or 20% below it was deemed to be an accurate reflection ideal weight for height, when compared to This study retrospectively analysed an of petrol sniffing history and the participant standard growth charts,21 only one criterion historical dataset, collected with the aim of was included in the study12 and furthermore, had to be met. investigating the social and neurological consistency between the consensual outcomes of adolescent inhalant abuse in assessment and self-assessment was high.18 Pooled groups Indigenous populations. Data collection All participants in the petrol sniffing group In initial studies using these participants; methods have been previously described met the criteria for inhalant abuse from the individuals with a history of petrol sniffing 9,12,17 in detail, but are summarised here and Diagnostic and Statistical Manual of Mental were separated based on the presence or discussed in the following paragraphs. Data Disorders IV. Drug use data were collected absence of lead encephalopathy. For this was collected from two remote Indigenous through an interview survey administered study, growth metrics were not different communities, where petrol sniffing had by the same researcher in conjunction with between these individuals, nor individuals been prevalent for many years. All subjects an Indigenous Research Assistant recruited between the two communities, and therefore were interviewed about petrol sniffing and from the same language group. This included individuals were not maintained in separate other drug use behaviour using a consensus age of onset/age petrol sniffing commenced groups for this study. To investigate the need methodology with survey instruments (years), duration (years), and severity (scale to control for the effects of other drug use developed specifically for this project, and of 1-5: 1/2=light/intermittent, 3=moderate, on growth, groups were stratified into the underwent a health check that included 4/5=heavy). The severity score was self-rated following; controls who use no other drugs neurological and cognitive examinations, based upon petrol sniffing behaviour in (n=15), controls who use one or two other collection of a blood sample, and height and the previous 12 months and was validated drug type use (n=13), controls who use three weight measurements. through a significant positive correlation or more other drugs (n=4) petrol sniffing with with the volume of petrol that participants no other drug use (n=9), petrol sniffing who 18 Participants reported having sniffed each week. use one or two other drugs (n=66), petrol Ethical approval for data collection was Participants were also surveyed regarding sniffing who use three or more other drugs obtained from the institutional ethics other drug use including alcohol, cigarette (n=11). These stratifications did not result committee, an independent Indigenous smoking, kava and marijuana. in any significant intra-group differences in ethics committee, and town councils from the height or weight (p>0.05). Thus individuals communities in the study. Ethical approval Haematological measures were pooled to form two groups; petrol for this analysis was granted by the Human Blood specimens were collected by sniffing and non-petrol sniffing control. Research Ethics Committee of Northern venepuncture. For those in the petrol sniffing Territory Department of Health and Menzies group, samples were collected within School of Health Research (HREC-2014-

2 Australian and New Zealand Journal of Public Health 2016 Online © 2016 Public Health Association of Australia Consequences of adolescent inhalant use

Data analysis Table 1: Age of commencement and severity of any drug use. Statistical analysis was conducted using IBM Non-petrol sniffing controls Petrol sniffing group p value SPSS Statistics 22 and power calculations Petrol sniffing (n) N/A 86 - were conducted using G*Power 3.1.9.2. Data Age commenced (years) N/A 12.9 ± 0.4 - for each variable was assessed for normality, Severity score (0-5) N/A 4.1 ± 0.4 - and normalised as necessary using square Cigarette smoking (n) 9 63 root transformation. Group differences were Presence of smoking (%) 28.1 73.3 0.000** investigated using independent sample Age commenced (years) 18.3 ± 1.5 15.3 ± 0.4 0.033* t-tests. The linear relationship between Severity score (0-5) 1.9 ± 0.3 2.4 ± 0.1 0.106 continuous variables was investigated Alcohol drinking (n) 15 58 using linear regression and 2-tailed Pearson Presence of alcohol drinking (%) 46.9 67.4 0.053^ Correlation coefficients. Chi-square tests were Age commenced (years) 19.3 ± 1.3 16.8 ± 0.4 0.055^ used to investigate the relationship between Severity score (0-5) 2.8 ± 0.4 2.9 ± 0.2 0.788 categorical variables. Significance was set at Kava (n) 1 21 p≤0.05; trends reported at p>0.05 but ≤0.10. Presence of kava use (%) 3.1 24.4 0.007** Results are reported as mean±SEM. Age commenced (years) 28.0 17.0 ± 0.9 0.012* Severity score (0-5) 1.0 2.5 ± 0.3 0.315 Marijuana (n) 7 41 Results Presence of marijuana use (%) 21.9 47.7 0.011* Petrol sniffing Age commenced (years) 22.0 ± 3.0 17.3 ± 0.7 0.084^ Severity score (0-5) 1.7 ± 0.4 2.3 ± 0.2 0.283 The age of commencement for petrol sniffing ^p>0.5 but ≤0.1; *p≤0.05; **p≤0.01. was 12.9±0.4 years. The average duration Groups were non-petrol sniffing controls (n=32) and petrol sniffing individuals (n=86). Group differences between the presence/absence of drug use (%) were was 8.6±0.8 years and severity score 4.1±0.4. assessed using chi-square tests. Group differences between age of commencement and severity score for each drug use were assessed using independent There were significant linear correlations sample t-tests (mean±SEM). There is no SEM for kava use in the non-petrol sniffing control group, as n=1 respondent. Petrol sniffing commenced earlier than other drug use and was associated with increased and earlier use of other drugs, but not with increased severity of other drug use. between all three metrics of petrol sniffing behaviour (Supplementary Table 2), with the Haematological data percentile (p=0.001) between petrol sniffing severity of petrol sniffing being predicted by (19th±2nd percentile) and non-petrol sniffing both duration and age of commencement Results for haematological variables are groups (39th±5th percentile) and in weight-for- (p=0.012, linear regression). There was also a shown in Supplementary Table 1. There was a age percentile (p=0.009, Figure 2). Increased significant correlation between the severity significant difference in serum toluene levels duration of petrol sniffing was linearly score and blood lead levels (p=0.014), (p<0.000) between petrol sniffing (0.06±0.02 associated with reduced height percentile confirming the validity of the severity score. µg/mL) and non-petrol sniffing groups (0.00±0.00 µg/mL). However, 73.5% of petrol (p=0.024, linear regression, Figure 3), but not weight percentile. Age of commencement Other drug use sniffing individuals had 0.00 µg/mL serum toluene. and severity of petrol sniffing were not Petrol sniffing was predominantly the correlated with either height or weight There was a significant difference in creatine first drug used, with 48.1% of all petrol percentiles. When stratified into sub-groups kinase levels (p=0.002) between petrol sniffing individuals reporting that as their by other drug use, there were no significant sniffing (420.2±39.3 U/L) and non-petrol first substance; cigarette smoking was the intra-group differences in height or weight. sniffing groups (212.7±22.4 U/L). There was a next most common first substance (6.5%). Furthermore, there was no significant significant group difference in exceeding the In comparison, cigarette smoking was difference between groups for BMI (Figure creatine kinase reference range (p=0.035, chi the most common first substance in the 1C) and BMI percentile. control group (17.6%); alcohol consumption square). Neither serum toluene nor creatine kinase levels were correlated with any of the the next most common (11.8%). Petrol Failure to thrive sniffing was associated with an increased three metrics of petrol sniffing behaviour Of the petrol sniffing group, 49% were less proportion of all other drug use (trend for (p>0.05). than 3rd percentile for weight and thus met alcohol, p=0.053) and with earlier other drug FTT criteria compared to 25% in the non- use, which was significant for the average Growth data sniffing group (p=0.022, chi square). Of the 48 age of commencement of cigarette and There was a significant difference in height individuals who met FTT criteria, 83% were kava use, with trends for earlier alcohol (Figure 1A) and weight (Figure 1B) between petrol sniffing individuals. (p=0.055) and marijuana (p=0.084) use petrol sniffing and non-petrol sniffing groups, with petrol sniffing individuals on average (Table 1). Furthermore, the age that sniffing Diagnosing petrol sniffing commenced was positively linearly correlated 5 cm shorter (p<0.000, with power of 0.95) with age of other drug use commencement and 7 kg lighter (p=0.001, with power of The existing diagnostic model of serum for cigarette smoking (p=0.022), kava 0.86). The regression co-efficient for weight toluene had a sensitivity of 27% and a (p=0.048) and marijuana (p=0.040), but not as predicted by height was 0.70 (p<0.000) in specificity of 100% with an overall accuracy of alcohol. The severity of other drug use was the petrol sniffing group and 0.84 (p=0.003) 47%, reflecting the high rate of false negatives not significantly different between petrol in the non-petrol sniffing group. There was with no false positives. Creatine kinase had sniffing and non-sniffing groups (Table 1). also a significant difference in height-for-age a sensitivity of 35% and a specificity of 84%

2016 Online Australian and New Zealand Journal of Public Health 3 © 2016 Public Health Association of Australia Crossin et al.

Figure 1: Petrol sniffing is associated with decreased (A) height (cm) and (B) weight (kg), but there was no change in (C) body mass index (BMI) (kg/m2).

Data are presented as minimum/maximum boxplots. n=32 for non-petrol sniffing controls and n=82 for petrol sniffing individuals, as not all participants had measurements for these variables. **p≤0.01. with overall accuracy of 49%. Using FTT as toluene and creatine kinase as diagnostic is associated with neurocognitive pathology a diagnostic model resulted in a sensitivity models of chronic inhalant abuse, and and impairment5 and is a risk factor for of 49% and a specificity of 75% with overall FTT had the highest sensitivity and overall ongoing psychological harm.23 A history accuracy of 56%, reflecting a lower rate of accuracy for chronic petrol sniffing, which of drug use also increases the risk of false negatives. represents a novel and practical diagnostic psychotic outcomes including suicide.24 tool for this behaviour. Thus adolescent inhalant abuse may have a Discussion In our cohort, petrol sniffing commenced in direct contribution to lifelong psychological early adolescence and was predominantly impacts, and also contribute indirectly by We explored the effects of chronic petrol the first substance misused; a consistent increasing subsequent drug use. These sniffing during adolescence on subsequent finding with other demographic studies findings highlight the need for early diagnosis drug use and growth in a cohort of in Australia and overseas.2,22 Additionally, and intervention of inhalant abuse, ideally Indigenous males. We expanded initial commencement age was positively before individuals’ progress to other drugs 18 observations from this dataset to show that correlated with severity and duration of of abuse. Petrol sniffing during adolescence petrol sniffing was associated with increased petrol sniffing. This relationship is important significantly impaired weight. In humans, and earlier onset of subsequent drug use, and as duration of petrol sniffing significantly adolescent inhalant abuse is associated 15 the commencement age of petrol sniffing increases the magnitude of neurological and with weight loss and emaciation. These also correlated positively with that of other cognitive deficits.12 The findings of increased findings are recapitulated in rodent studies drug use. Petrol sniffing also impaired both and earlier drug use after petrol sniffing are and associated with reduced food intake in height and weight, and weight impairments consistent with other studies suggesting the absence of anhedonia, decreased weight 14 were so severe that half the petrol sniffing that inhalant use appears to establish a gain and reduced fat deposition. In humans individuals met FTT diagnosis. In this cohort, trajectory towards other drug use (see for there is evidence that incidental inhalation growth impairment out-performed serum review Dinwiddie 1994).11 This is of significant of petrol fumes may have an acute anorectic 25 concern as increased drug use in adolescence effect, suggesting that weight impairment Figure 2: Weight percentile difference between may be driven by decreased food intake, groups, presented as minimum/maximum boxplots while others suggest that toluene exposure Figure 3: Increased duration of petrol sniffing 26 with line on y-axis at 3rd percentile, denoting results in a higher basal metabolic rate or behaviour (years) is correlated with decreased 14 meeting failure to thrive criteria. alters appetite regulating hormones. Body height percentile (p=0.024), n=38 due to not all weight is driven by a complex relationship respondents providing data on duration of petrol between food intake, metabolic function, sniffing behaviour. body composition and energy expenditure;27 our dataset does not allow mechanistic interpretation. Nevertheless, the observed weight impairments were so severe that nearly 50% of petrol sniffing individuals met FTT criteria. FTT is not a disease, but rather a symptomatic description of poor growth attributable to many causes, including malnutrition or psychosocial factors including poverty.28 Whilst the baseline prevalence Petrol sniffing is associated with decreased weight percentile (p=0.009). A of FTT is higher in Indigenous populations break in the y axis below the 10th percentile is utilised to clearly display 29 left-skewed data. n=32 for non-petrol sniffing controls and n=82 for Data are presented as a scatter plot with a calculated regression line compared to non-Indigenous populations, petrol sniffing individuals. **p≤0.01. showing the relationship between the variables. we identified differences in the FTT rates

4 Australian and New Zealand Journal of Public Health 2016 Online © 2016 Public Health Association of Australia Consequences of adolescent inhalant use

within these two communities, based upon facility, given their requirement for blood historic cohort of Indigenous males from a history of petrol sniffing in adolescence. collection. Since inhalant abuse is associated two remote communities. However, data Importantly, childhood FTT is associated with low socioeconomic status and inhalant from human inhalant abuse is difficult with an increased risk of subsequent abusers are disproportionately represented to obtain and, as drug use, growth and disease development, including obesity and in the mental health and protective services FTT are common measures of health and cardiovascular disease.30Adolescent petrol systems, these populations have less development, this study provides relevant sniffing also significantly impaired height, a regular contact with the medical system.7 In observations. One limitation of this study novel finding. Height was also a significant addition, petrol sniffing among Indigenous is that birth weight data was not available; predictor of weight in both petrol sniffing and people occurs predominantly in remote in order to assess growth trajectories prior non-petrol sniffing individuals, suggesting communities, where access to health services to petrol sniffing. However, the use of a that the weight impairments may be partially is limited. Therefore we investigated whether control group from the same communities attributable to impaired height. Adolescence the effects of inhalants on growth could be in this study provides some mitigation encompasses an intensive period of growth incorporated into a novel diagnostic tool for for this potential confound. Furthermore, (adolescent growth spurt) where over half of identifying chronic inhalant abuse. In our while low socioeconomic status and the peak bone mass is attained.31 For males, study, height percentile was significantly disadvantage are risk factors for FTT, and the growth spurt initiates around 12 years impaired by petrol sniffing and importantly, are associated with inhalant use, the control of age and ends at about 20 years of age,32 was also negatively correlated with increased group also provides mitigation for the risk which coincides with the primary time of duration of petrol sniffing behaviour, of these factors confounding our findings. petrol sniffing behaviour in this study. As this suggesting this metric may be sensitive to In addition, our ability to extrapolate these growth spurt can be disrupted by factors such identifying the nuances of chronic abuse. findings to females or to urban populations as malnutrition33 it is likely that inhalant abuse Furthermore, individuals with a history is limited. There is also a potential confound could disrupt this intensive growth period, of petrol sniffing met FTT criteria more of lead in this cohort, as individuals sniffed including skeletal growth. Indeed chronic frequently than non-sniffers. Thus growth a combination of leaded and unleaded toluene exposure impairs both skeletal metrics may be a clinically relevant tool for petrol, which has also been associated with growth and bone mineral density in rats16,34 identifying chronic adolescent inhalant abuse impaired weight and height.41 However, and bone mineral density in adolescent males having higher sensitivity and overall accuracy we suggest that these findings are relevant with a history of chronic glue sniffing.35 Any than any of the existing diagnostic tools. In to other inhalants, as toluene is a common disruption to growth during adolescence addition, growth metrics were almost twice solvent in all inhaled products and animal has the potential to cause long-term effects as sensitive as serum toluene in identifying studies using toluene have shown similar on stature.36 As both height and weight chronic inhalant abuse. For a screening test, growth impairments as in this study.14,16 were reduced, there was no change in BMI sensitivity is more valuable than specificity, as While further work will be needed to identify between petrol sniffing and non-petrol the focus for inhalant abuse needs to be on the mechanisms underlying these growth sniffing groups. This highlights the risk of improving the identification of a serious but impairments, particularly the effect of using BMI as a primary metric of growth. treatable condition. inhalant-induced appetite suppression, the Current diagnostic tools for inhalant abuse The use of growth metrics as a diagnostic tool persistence of these effects into abstinence, are limited, and restricted to presentation to is feasible, given that body growth is routinely and the health consequences of growth a medical facility under acute intoxication, measured by an individual or a parent/ impairment during adolescence, we suggest and the subsequent measurement of serum guardian, it does not change rapidly and that height impairment should be added to toluene or creatine kinase levels. In this measurement does not require contact with advisory notes as a warning sign of inhalant study, 73.5% of the petrol sniffing individuals the primary health care system or specialised abuse. had a zero reading of serum toluene within equipment. This may be especially valuable This is the first study that has established a 12 hours of petrol sniffing. Creatine kinase for disadvantaged and remote populations. It link between FTT and adolescent inhalant levels were elevated by petrol sniffing and is unknown how quickly growth is impaired abuse, potentially providing a novel may provide a more temporally stable by inhalant abuse; therefore, this approach diagnostic technique for chronic inhalant measure than serum toluene.9 However, the is more likely to be applicable to chronic abuse thus increasing the opportunity for relevance of creatine kinase to inhalants use. Growth impairments arising from intervention. Early intervention is effective other than petrol is unknown. Furthermore, adolescent inhalant abuse may also provide in relation to petrol sniffing as cognitive creatine kinase levels can be increased by the basis of a powerful health education impairments can resolve with sustained other environmental factors such as high message. Evidence suggests that some abstinence.17 Furthermore, early intervention ambient temperature37 or other drug use.38 inhalant users are motivated by a desire to can reduce subsequent drug use, adverse Neither serum toluene nor creatine kinase restrict their weight;39 however, it may be less psychological outcomes and other high-risk correlated with severity or duration of petrol desirable to also restrict height. This health behaviours.42 We suggest that inhalant abuse sniffing behaviour and were therefore not promotion message may also be relevant to should be added to the differential diagnosis helpful metrics for diagnosing chronic the study group we investigated, adolescent guidance for FTT in high risk populations and inhalant abuse. Indigenous males, where size and strength age groups. 40 Importantly, the measures indicated above are highly valued personal attributes. Our require acute presentation to a medical data were retrospectively analysed from a

2016 Online Australian and New Zealand Journal of Public Health 5 © 2016 Public Health Association of Australia Crossin et al.

6. Kurtzman TL, Otsuka KN, Wahl RA. Inhalant abuse by 27. Woods SC, Seeley RJ, Porte D, Schwartz MW. Signals that Conclusion adolescents. J Adolesc Health. 2001;28(3):170-80. regulate food intake and energy homeostasis. Science. 7. Kang M, Bernard D, Booth M, Quine S, Alperstein G, 1998;280(5368):1378-83. This study shows that petrol sniffing is Usherwood T, et al. Access to primary health care for 28. Shah MD. Failure to thrive in children. J Clin Gastroenterol. typically the first substance misused and is Australian young people: Service provider perspectives. 2002;35(5):371-4. associated with increased and earlier use of Br J Gen Pract. 2003;53(497):947-52. 29. Northern Territory Department of Health and Families. 8. Meulenbelt J, De Groot G, Savelkoul T. Two cases of acute Child Malnutrition / Failure to Thrive. Alice Springs other drugs. This misuse commences during toluene intoxication. Br J Ind Med. 1990;47(6):417-20. (AUST): Government of Northern Territory; 2008. a critical period for maturational processes. 9. Burns CB, Powers JR, Currie BJ. Elevated serum creatine 30. Stein AD, Thompson AM, Waters A. Childhood kinase (CK-MM) in petrol sniffers using leaded or growth and chronic disease: Evidence from countries Petrol sniffing significantly impaired weight unleaded fuel. Clin Toxicol. 1994;32(5):527-39. undergoing the nutrition transition. Matern Child Nutr. and height, independent of other drug use, 10. Lubman DI, Yücel M, Lawrence AJ. Inhalant abuse 2005;1(3):177-84. among adolescents: Neurobiological considerations. 31. Bonjour J-P, Theintz G, Buchs B, Slosman D, Rizzoli to the extent that many individuals met the Br J Pharmacol. 2008;154(2):316-26. R. Critical years and stages of puberty for spinal and criteria for a FTT diagnosis. In combination, 11. Dinwiddie SH. Abuse of inhalants: A review. Addiction. femoral bone mass accumulation during adolescence. these findings suggest that adolescent 1994;89(8):925-39. J Clin Endocrinol Metab. 1991;73(3):555-63. 12. MaruffP, Burns C, Tyler P, Currie B, Currie J. Neurological 32. Tanner JM. Growth and maturation during adolescence. inhalant abuse can initiate a trajectory of and cognitive abnormalities associated with chronic Nutr Rev. 1981;39(2):43-55. substance misuse and growth impairment, petrol sniffing. Brain. 1998;121(10):1903-17. 33. Dreizen S, Spirakis CN, Stone RE. A comparison of 13. Yip PS, Liu K-Y, Lam T, Stewart SM, Chen E, Fan S. skeletal growth and maturation in undernourished and thus highlights the need for early Suicidality among high school students in Hong Kong, and well-nourished girls before and after menarche. J diagnosis. Using growth parameters as SAR. Suicide Life Threat Behav. 2004;34(3):284-97. Pediatr. 1967;70(2):256-63. 14. Dick A, Simpson A, Qama A, Andrews Z, Lawrence 34. Atay AA, Kismet E, Turkbay T, Kocaoglu M, Demirkaya a diagnostic tool for inhalant abuse may A, Duncan J. Chronic intermittent toluene inhalation E, Sarici SU, et al. Bone mass toxicity associated with be clinically valuable as they are simple in adolescent rats results in metabolic dysfunction inhalation exposure to toluene. Biol Trace Elem Res. to implement and can be translated into with altered glucose homeostasis. Br J Pharmacol. 2005;105(1-3):197-203. 2015;172(21):5174-87. 35. Dündaröz MR, Sarici S, Turkbay T. Evaluation of bone populations where inhalant abuse is more 15. Glaser HH, Massengale ON. Glue-sniffing in children: mineral density in chronic glue sniffers. Turk J Pediatr. prevalent. Given the current difficulties Deliberate inhalation of vaporized plastic cements. 2002;44:326-9. JAMA. 1962;181(4):300-3. 36. Martorell R, Khan LK, Schroeder DG. Reversibility of in identifying inhalant misuse, our study 16. Pryor GT. A toluene-induced motor syndrome in rats stunting: Epidemiological findings in children from provides a novel opportunity for improving resembling that seen in some human solvent abusers. developing countries. Eur J Clin Nutr. 1994;48:S45-57. Neurotoxicol Teratol. 1991;13(4):387-400. 37. Knochel JP, Dotin LN, Hamburger RJ. Heat stress, diagnostic capabilities thus leading to 17. Cairney S, Maruff P, Burns CB, Currie J, exercise, and muscle injury: Effects on urate metabolism earlier intervention, in order to alter the Currie BJ. Neurological and cognitive recovery and renal function. Ann Intern Med. 1974;81(3):321-8. trajectory of ongoing drug use and prevent following abstinence from petrol sniffing. 38. Swartz CM, Breen KJ. Elevated serum CK in long Neuropsychopharmacology. 2005;30(5):1019-27. abstinent abusers. Am J Drug Alcohol Abuse. chronic adverse psychological and health 18. Burns C, d’Abbs P, Currie B. Patterns of petrol sniffing 1993;19(3):327-35. consequences. and other drug use in young men from an Australian 39. Pisetsky EM, May Chao Y, Dierker LC, May AM, Aboriginal community in Arnhem Land, Northern Striegel‐Moore RH. Disordered eating and substance Territory. Drug Alcohol Rev. 1995;14(2):159-69. use in high‐school students: Results from the Youth Acknowledgements and funding 19. Centre for Disease Control. Growth Charts, United States, Risk Behavior Surveillance System. Int J Eat Disord. 2000. Atlanta (GA): National Center for Health Statistics 2008;41(5):464-70. The authors thank Dr Chris Burns and National Center for Chronic Disease Prevention and 40. Mellor D, McCabe M, Ricciardelli L, Ball K. Body Professor Paul Maruff for data collection. Health Promotion; 2000. image importance and body dissatisfaction among 20. Fryar CD, Gu Q, Ogden CL. Anthropometric reference Indigenous Australian adolescents. Body Image. The research was supported by NHMRC data for children and adults: United States, 2007-2010. 2004;1(3):289-97. (940835), of which AJL is a Principal Research Vital Health Stat 11. 2012;(252):1-48. 41. Schwartz J, Angle C, Pitcher H. Relationship between 21. Perrin EC, Cole C, Frank D, Glicken SR, Guerina N, Petit childhood blood lead levels and stature. Pediatrics. Fellow (1020737), the Australian Research K, et al. Criteria for determining disability in infants and 1986;77(3):281-8. Council (DP 110100379) of which JRD was a children. Evid Rep Technol Assess (Summ). 2003;(72):1-5. 42. Toumbourou JW, Stockwell T, Neighbors C, Marlatt Future Fellow during the time of the study 22. Dell CA, Gust SW, MacLean S. Global issues in volatile G, Sturge J, Rehm J. Interventions to reduce harm substance misuse. Subst Use Misuse. 2011;46 Suppl associated with adolescent substance use. Lancet. (100100235) and the Victorian Government’s 1:1-7. 2007;369(9570):1391-401. Operational Infrastructure Support Scheme. 23. Armstrong TD, Costello EJ. Community studies on adolescent substance use, abuse, or dependence and psychiatric comorbidity. J Consult Clin Psychol. 2002;70(6):1224. Supporting Information References. 24. Borges G, Walters EE, Kessler RC. Associations Additional supporting information may be 1. Alcohol and other Drugs Council of Australia. Policy of substance use, abuse, and dependence with Position - Inhalants. Canberra (AUST): ADCA; 2010. subsequent suicidal behavior. Am J Epidemiol. found in the online version of this article: 2. Australian Institute of Health and Welfare. 2010 National 2000;151(8):781-9. Drug Strategy Household Survey Report. Canberra 25. Jackson CE, Currie BJ, Cairney S, Maruff PT, Snyder Supplementary Table 1: Methods and (AUST): AIHW; 2011. PJ. Hunger and the perception of the scent of petrol: results of haematological variables. 3. Cairney S, Maruff P, Burns C, Currie B. The A potential neurobiological basis for increased neurobehavioural consequences of petrol (gasoline) risk of petrol inhalation abuse. Addict Res Theory. Supplementary Table 2: Relationship sniffing.Neurosci Biobehav Rev. 2002;26(1):81-9. 2009;17(5):518-24. between metrics of petrol sniffing behaviour. 4. Cancer Council of Victoria. Australian Secondary School 26. Wang D-H, Ishii K, Seno E, Yane S, Horike T, Yamamoto Students’ Use of Tobacco, Alcohol, and Over-the-counter H, et al. Reduced serum levels of ALT and GGT and and Illicit Substances in 2011. Melbourne (AUST): high carbohydrate intake among workers exposed to Australian Department for Health and Ageing; 2012. toluene below the threshold limit values. Ind Health. 5. Lubman DI, Yücel M, Hall WD. Substance use 1998;36(1):14-9. and the adolescent brain: A toxic combination? J Psychopharmacol. 2007;21(8):792-4.

6 Australian and New Zealand Journal of Public Health 2016 Online © 2016 Public Health Association of Australia Consequences of adolescent inhalant abuse

Supplementary Table 1 – Methods and results of haematological variables

Result (mean±SEM) Variable Unit A measure of Method of analysis Non-sniffing Petrol sniffing p value controls individuals Hemoglobin g/L Complete blood count Coulter S+4 counter 146.8±2.0 141.4±2.0 0.114 Red cell count 106/µL Complete blood count Coulter S+4 counter 5.2±0.1 5.1±0.1 0.299 Hematocrit / packed L/L Complete blood count Coulter S+4 counter 0.422±0.005 0.414±0.005 0.386 cell volume Mean cell volume fL Complete blood count Coulter S+4 counter 80.8±0.7 80.8±0.7 0.920 Red cell distribution Percentage Complete blood count Coulter S+4 counter 13.8±0.3 14.1±0.2 0.433 width Platelets 109/L Complete blood count Coulter S+4 counter 274.3±14.1 255.0±8.6 0.239 White cell count 109/L Complete blood count Coulter S+4 counter 9.3±0.4 9.2±0.3 0.778 Kodak “dry slide” Creatine kinase U/L Muscle inflammation 212.7±22.4 420.2±39.3 0.002 ** technique Smooth muscle Kodak “dry slide” Creatine kinase B U/L 1.8±0.4 3.0±0.3 0.075^ inflammation technique Iron µmol/L Iron panel Kodak E 700 14.5±0.8 16.2±0.8 0.202 Transferrin g/L Iron panel Beckman array 3.3±0.1 3.5±0.1 0.094^ Transferrin saturation % Calculation (% transferrin Iron panel 0.22±0.02 0.22±0.01 0.970 rate Saturation saturated with iron) Ferritin µg/L Iron panel Baxter Stratus 89.4±10.4 70.1±6.7 0.126 Blood lead µM/L Petrol exposure Standard additions method 0.27±0.03 1.52±0.10 0.000 ** Gas chromatograph mass Blood toluene µg/mL Petrol exposure 0.00±0.00 0.06±0.02 0.012 * spectrometry Gas chromatograph mass Blood benzene µg/mL Petrol exposure 0.000±0.000 0.011±0.003 0.057^ spectrometry

1

Consequences of adolescent inhalant abuse

The group differences between non-petrol sniffing controls and petrol sniffing individuals were measured using independent sample t-tests. For all haematological variables n=115 (32 controls and 83 petrol sniffing individuals). Results are presented as mean±SEM for each group. ^ p>0.5 but ≤0.1; * p≤0.05; ** p≤0.01.

2

Consequences of adolescent inhalant abuse

Supplementary Table 2 – relationship between metrics of petrol sniffing behaviour

Metric Age sniffing Duration of Severity of commenced sniffing sniffing Age sniffing commenced (years) 1 -0.426 -0.254 (0.008)** (0.040)* Duration of sniffing (years) -0.426 1 0.486 (0.008)** (0.003)** Severity of sniffing (scale 1-5) -0.254 0.486 1 (0.040)* (0.003)**

Only individuals in the petrol sniffing group (n=86) were asked this question.

Participation in all questions were optional and for this question n=72 for age sniffing commenced, n=66 for severity, and n=40 for duration. Results are reported as a two-tailed

Pearson’s correlation coefficient with associated p value in brackets. There is a significant correlation between all three metrics of petrol sniffing behaviour. *p≤0.05; **p≤0.01.

ADDICTION RESEARCH & THEORY, 2017 https://doi.org/10.1080/16066359.2017.1339229

RESEARCH ARTICLE The persistence of growth impairments associated with adolescent inhalant abuse following sustained abstinence

Rose Crossina , Sheree Cairneyb,c,d, Andrew John Lawrencea,e and Jhodie Rubina Duncana,f aAddiction Neuroscience, Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia; bCentre for Remote Health, Flinders University, Alice Springs, NT, Australia; cCooperative Research Centre for Remote Economic Participation (CRC-REP), Ninti One Limited: Information-Innovation-Ideas for Remote Australia, Alice Springs, NT, Australia; dMenzies School of Health Research, Casuarina, NT, Australia; eFlorey Department of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC, Australia; fSchool of Medicine, University of Adelaide, Adelaide, SA, Australia

ABSTRACT ARTICLE HISTORY Background: Abuse of inhalants containing the volatile solvent toluene is a significant public health Received 8 January 2017 issue, especially for adolescent and Indigenous communities. We previously demonstrated that inhalant Revised 1 June 2017 abuse (petrol sniffing) during adolescence results in impairments to height and weight. The aim of this Accepted 3 June 2017 study was to understand whether these impairments resolve or persist into early adulthood, following sustained abstinence. KEYWORDS Methods: Baseline data were collected from 118 Indigenous males; 86 chronically sniffed petrol during Petrol sniffing; height; adolescence. Following 2 years sustained abstinence, data were again collected from a subset (n ¼ 40) weight; Indigenous health; of this population; 30 sniffed petrol during adolescence. This study is a retrospective analysis of data volatile solvent abuse; body collected after 2 years sustained abstinence. mass index Results: After 2 years abstinence, inhalant-induced impairments to height persisted (p ¼ 0.023) whereas weight impairments resolved (p ¼ 0.796). Conclusions: Adolescent inhalant abuse alters growth trajectories, even after 2 years of sustained abstinence. Despite the fact that individuals continue to get taller, there is no catch-up growth in those who abused inhalants. The persistence of height impairments demonstrates that adolescent inhalant abuse can impact individuals into adulthood, despite sustained abstinence. In contrast, weight impair- ments associated with inhalant abuse resolved in abstinence, however, it is unknown if this represents a normalisation of weight or a rapid and unhealthy gain in weight. Further research is required to determine the health impacts of the observed weight changes.

Abbreviations: BMI: Body mass index

Introduction (Bonjour et al. 1991). Therefore, an impact on height has the potential to affect growth trajectories into adulthood (Reed Inhalant abuse is the deliberate inhalation of solvents (e.g. & Stuart 1959). Additionally, dramatic weight fluctuations petrol or glue), to achieve an altered mental state. can adversely impact health (Norgan 2000; Dulloo et al. Adolescents comprise the predominant population of inhal- 2006). Therefore, we investigated whether our observed ant abusers (AIHW 2011). In human and/or animal studies, weight and height changes arising from adolescent inhalant appetite suppression, reduced weight gain (Glaser & abuse (Crossin et al. 2016) persist following abstinence; cur- Massengale 1962; Dick et al. 2015) and depressed skeletal rently an open question. growth (Pryor 1991) are reported consequences of adolescent inhalant abuse. We previously showed that petrol sniffing during adolescence impairs height and weight, with petrol Methods sniffing individuals on average 5 cm shorter and 7 kg lighter than non-petrol sniffing controls (Crossin et al. 2016). This We undertook retrospective analysis of a historical dataset, study was the first to show height impairments arising from collected from a cohort of Indigenous males from a remote chronic inhalant abuse during adolescence (Crossin et al. northern Australian community. The current study utilises 2016). data collected as a follow-up after two years sustained abstin- Despite awareness of acute consequences, the long-term ence from chronic petrol sniffing (a combination of leaded effects of adolescent inhalant abuse on growth remain and unleaded petrol), achieved through a community inter- unknown but require consideration, as the peak time of vention programme. Data collection methods and validation inhalant abuse coincides with the adolescent growth spurt of the community-led intervention have been previously

CONTACT Rose Crossin Florey [email protected] Institute of Neuroscience and Mental Health University of Melbourne, Parkville, Victoria 3052, Australia ß 2017 Informa UK Limited, trading as Taylor & Francis Group. 2 R. CROSSIN ET AL. described (Burns et al. 1995a; Maruff et al. 1998), and are there were no volatile hydrocarbons detected in the blood of summarised below. any participants after two years abstinence and no differen- ces were detected in creatine kinase levels between the Participants groups (Burns et al. 1995a). The use of cigarettes, alcohol, and remained consistent between the two time Ethical approval for this analysis was granted by the points, with a significant reduction in kava use after two Human Research Ethics Committee of Northern Territory years (Burns et al. 1995a). Department of Health and Menzies School of Health Research (HREC-2014-2274). Informed written consent was Growth measures given by all participants, who could withdraw from the study at any time. Height (cm) and weight (kg) were measured by the research We previously described the metrics of the historic petrol team during the interview and body mass index (BMI) cal- sniffing behaviour (Crossin et al. 2016). In summary, the age culated (weight (kg)/height squared (m2)). Weight was meas- of commencement for petrol sniffing was 12.9 ± 0.4 years. ured on an electronic scale and height was measured using a The average duration was 8.6 ± 0.8 (minimum 1, maximum tape. These measures were undertaken by a single trained 21) years and severity score 4.1 ± 0.4 (scale from 0-5, 5 indi- individual to prevent inter-rater variability, replicating previ- cating heaviest use) and the initial study was conducted at ous methodology (Crossin et al. 2016). an average age of 20.6 ± 0.4 years (Crossin et al. 2016). When data were originally collected, petrol sniffing was quantified as it related to a 375 mL soft drink can, as well as the sever- Data analysis ity score. The mean volume of petrol sniffed was 250 mL per Statistical analysis was conducted using IBM SPSS Statistics night and there was a significant correlation (p < 0.001) 22. Data for each variable was assessed for normality, and between the quantification estimate and the severity score normalised as necessary using square root transformation. (Burns et al. 1995b). Therefore the severity score was then Group differences were investigated using independent sam- used as the primary measure of consumption for the ple t-tests, which have high accuracy with small sample sizes. analysis. Data presented as mean ± SEM. Significance was set at After two years confirmed abstinence from petrol sniffing, p 0.05, trends reported at p > 0.05 0.1. 40 participants, a sub-set of the original study cohort, were re-interviewed by the original research team and an Indigenous Research Assistant recruited from the same lan- Results guage group. 30 participants had a history of petrol sniffing in adolescence, 10 non-petrol sniffing age-matched individu- Growth data is summarised in Table 1. After two years als acted as controls. Whilst the initial study was conducted abstinence, individuals with a history of petrol sniffing (age across two communities, only one community was revisited 23.0 ± 0.7) were on average 6 cm shorter than non-petrol for the follow-up study at two years post-abstinence. Within sniffing controls (age 22.3 ± 1.5). In contrast, there was no that community the retention rate in the study was 40 of the significant difference in body weight or BMI between the 58 initial participants, with the predominant reason for two groups, though there was a trend towards increased per- dropping out of the study being that individuals had left the centage weight gain in petrol sniffing individuals. community in the intervening two years (Burns et al. 1995a). Abstinence was confirmed by self-assessment combined Discussion with a consensual methodology, cross-checked against hos- pital admission records and confirmed by Aboriginal health We explored the persistence of growth impairments arising workers (Burns et al. 1995a; Cairney et al. 2005). Abstinence from inhalant abuse in a cohort of Indigenous males who was further biochemically confirmed by the finding that chronically sniffed petrol during adolescence, followed by

Table 1. Growth data after two years abstinence. After two years abstinence Non-petrol sniffing controls Petrol sniffing individuals p value Age (years) 22.3 ± 1.5 23.0 ± 0.7 0.693 Height (cm) 177.0 ± 3.1 171.3 ± 1.0 0.023 Change in height (cm) 3.1 ± 0.8 3.8 ± 0.7 0.590 Change in height (%) 1.8 ± 0.5 2.3 ± 0.5 0.514 Weight (kg) 66.0 ± 6.1 64.6 ± 2.4 0.796 Change in weight (kg) 4.8 ± 3.3 9.2 ± 1.6 0.191 Change in weight (%) 7.2 ± 4.6 18.0 ± 3.4 0.100# BMI (kg/m2) 20.9 ± 1.6 22.0 ± 0.7 0.502 Change in BMI (kg/m2) 0.7 ± 1.0 2.3 ± 0.6 0.172 Change in BMI (%) 3.4 ± 4.0 12.9 ± 3.4 0.139 Height was reduced by an average of 6 cm in individuals who sniffed petrol during adolescence, whereas weight was not significantly different. n ¼ 10 for non-petrol sniffing controls and n ¼ 30 for petrol sniffing individuals. Data presented as mean ± SEM. #p > 0.05 0.1. p < 0.05. ADDICTION RESEARCH & THEORY 3 two years sustained abstinence. In this cohort we previously et al. 2013), opioids (Thornhill et al. 1976), methampheta- showed that petrol sniffing during adolescence was associ- mine (Williams et al. 2004) and inhalants (Crossin et al. ated with height impairment (Crossin et al. 2016). While 2016). This weight loss is often attributed to poor nutrition acknowledging the low sample size of the participant subset and appetite suppression (Islam et al. 2002; Petersson et al. in our follow up study as a potential limitation, our data 2004). However, many studies, in both animals and humans, show that the effects of inhalant abuse on height persist into have observed that the degree of weight loss is not directly early adulthood, despite sustained abstinence. Despite indi- comparable to measured changes in food intake, which sug- viduals in both the control and petrol sniffing groups con- gests that nutrition is not the sole mediator of drug-induced tinuing to get taller, there is no catch-up growth in those weight loss (Perkins 1992; Cooper & Van der Hoek 1993; who abused inhalants, suggesting that adolescent petrol sniff- Ersche et al. 2013). ing may alter height trajectory. The mechanism for inhalant- In contrast to active substance abuse, recovery/abstinence induced growth impairment is unknown, though one have a strong association with weight gain (Jackson & Grilo possible explanation is that growth impairments are a result 2002; Hodgkins et al. 2004), and our results are consistent of malnutrition, as inhalant abuse is associated with altered with this. Various hypotheses exist to explain this abstinence metabolism (Dick et al. 2015) and reduced food intake effect include; a homeostatic rebound effect to normalise (Moron et al. 2004). Alternatively, inhalant abuse throughout body weight (Nolan 2013), addiction switching from a drug the adolescent growth spurt may directly affect bone growth to palatable food (Brunault et al. 2015), or common reward- (Martorell et al. 1994). Regardless of the mechanism, persist- seeking phenotype between drugs and palatable food (Nolan ent impairments to height may have detrimental consequen- & Stolze 2012). However, there are limits to all these hypothe- ces including adverse psychological outcomes e.g. social ses and all focus on food intake as the mediator of the growth anxiety and poor self-image (Molinari et al. 2002), as well as changes. Further research is needed into the cause of weight adverse developmental and educational outcomes (Siegel regain after substance abuse before effective interventions can et al. 1990; Norgan 2000). be developed, to assist in the prevention of health issues such In contrast to height, there was no long term difference as insulin resistance (Dulloo et al. 2006) in abstinence. in body weight; however the percent weight change in the In conclusion, we show that petrol sniffing associated petrol sniffing group was more than double that of the non- height impairments persist into adulthood despite sustained petrol sniffing controls. It is unlikely that the rapid weight abstinence. This highlights the need for prevention and early gain during abstinence is due to withdrawal symptoms from intervention for adolescent inhalant abusers, as in contrast petrol sniffing, as these are characterised by anhedonia to cognitive and neurological consequences (Cairney et al. rather than increased food intake, and are transient (Shah 2005), abstinence alone cannot ameliorate the adverse et al. 1999), though we acknowledge that no data on with- growth consequences. Indeed, this height restricting effect drawal symptoms was collected as part of this study. Weight may present the opportunity for a prevention message about regain after weight loss is a common physiological response inhalant abuse that will be relevant to adolescents, for whom to re-establish homeostasis (Sumithran & Proietto 2013). size and strength are highly valued personal attributes However, rapid weight regain is associated with adverse con- (Mellor et al. 2004). We also show that abstinence from pet- sequences including increased fat deposition and the devel- rol sniffing results in a rapid weight gain, the cause of which opment of insulin resistance (Dulloo et al. 2006). is currently unknown, but has the potential to lead to further Furthermore, whilst the average BMI of study participants adverse health outcomes even though inhalant abuse behav- ‘ ’ was in the normal range after two years abstinence; it can- iours have ceased. not be assumed that a normal BMI indicates metabolic health. Individuals with a history of petrol sniffing may pre- sent as metabolically-obese normal-weight, a harmful pheno- Acknowledgements type associated with increased cardiovascular and diabetes The authors thank Dr Chris Burns and Professor Paul Maruff for data disease mortality (Carnethon et al. 2012) though this collection. The research was supported by NHMRC (940835), of which requires confirmation. A further potential confound is that AJL is a Principal Research Fellow (1020737), the Australian Research whilst individuals were in abstinence from petrol sniffing, Council (DP 110100379) of which JRD was a Future Fellow during the their use of other drugs such as cigarettes, alcohol, and mari- time of the study (100100235) and the Victorian Government’s juana continued, though these drugs were not previously Operational Infrastructure Support Scheme. Funding bodies had no involvement in the design, analysis and decision to publish. There are found to be predictors of growth outcomes in this cohort no conflicts of interest or financial disclosures in this work. (Crossin et al. 2016). Our results highlight the need for fur- ther research into the effects of inhalant abuse on body weight, particularly in abstinence, as body weight can signifi- Disclosure statement cantly influence long-term health outcomes (Dulloo et al. No potential conflict of interest was reported by the authors. 2006). Additional human cohorts with larger sample sizes would be valuable in this regard. The effect of petrol sniffing, and indeed, substance abuse Funding in general on body weight is poorly understood. Active sub- The research was supported by NHMRC (940835), of which AJL is a stance abuse is associated with impaired body weight for Principal Research Fellow (1020737), the Australian Research Council many different drug types, including cigarettes (Grebenstein (DP 110100379) of which JRD was a Future Fellow during the time of 4 R. CROSSIN ET AL. the study (100100235) and the Victorian Government’s Operational Hodgkins CC, Cahill KS, Seraphine AE, Frostpineda K, Gold MS. 2004. Infrastructure Support Scheme. Adolescent drug addiction treatment and weight gain. J Addict Dis. 23:55–65. Islam SN, Hossain KJ, Ahmed A, Ahsan M. 2002. Nutritional status ORCID of drug addicts undergoing detoxification: prevalence of malnutrition and influence of illicit drugs and lifestyle. BJN Nutr. Rose Crossin http://orcid.org/0000-0003-1814-1330 88:507–513. Jackson T, Grilo C. 2002. Weight and eating concerns in outpatient men and women being treated for substance abuse. Eat Weight Disord Obesity. 7:276–283. References Martorell R, Khan LK, Schroeder DG. 1994. Reversibility of stunting: epidemiological findings in children from developing countries. Eur AIHW. 2011. 2010 National Drug Strategy Household Survey Report. J Clin Nutr. 48:S45–S57. Canberra. Maruff P, Burns C, Tyler P, Currie B, Currie J. 1998. Neurological and Bonjour J, Theintz G, Buchs B, Slosman D, Rizzoli R. 1991. Critical cognitive abnormalities associated with chronic petrol sniffing. Brain. years and stages of puberty for spinal and femoral bone mass accu- 121:1903–1917. mulation during adolescence. J Clin Endocrinol Metabol. Mellor D, McCabe M, Ricciardelli L, Ball K. 2004. Body image import- 73:555–563. ance and body dissatisfaction among Indigenous Australian adoles- Brunault P, Salame E, Jaafari N, Courtois R, Reveillere C, Silvain C, cents. Body Image. 1:289–297. Benyamina A, Blecha L, Belin D, Ballon N. 2015. Why do liver Molinari E, Sartorio A, Ceccarelli A, Marchi S. 2002. Psychological and transplant patients so often become obese? The addiction transfer emotional development, intellectual capabilities, and body image in hypothesis. Med Hypotheses. 85:68–75. short normal children. J Endocrinol Invest. 25:321–328. Burns C, d'Abbs P, Currie B. 1995a. Patterns of petrol sniffing and Moron L, Pascual J, Portillo P, Casis L, Macarulla MT, Abecia LC, other drug use in young men from an Australian Aboriginal com- Echevarrı E. 2004. Toluene alters appetite, NPY, and galanin immu- munity in Arnhem Land, Northern Territory. Drug Alcohol nostaining in the rat hypothalamus. Neurotoxicol Teratol. Rev.14:159–169. 26:195–200. Burns CB, Currie BJ, Clough AB, Wuridjal R. 1995b. Evaluation of Nolan LJ. 2013. Shared urges? The links between drugs of abuse, eating, strategies used by a remote aboriginal community to eliminate petrol – sniffing. Med J Australia. 163:82–86. and body weight. Curr Obes Rep. 2:150 156. Nolan LJ, Stolze MR. 2012. Drug use is associated with elevated food Cairney S, Maruff P, Burns CB, Currie J, Currie BJ. 2005. Neurological – and cognitive recovery following abstinence from petrol sniffing. consumption in college students. Appetite. 58:898 906. Neuropsychopharmacology. 30:1019–1027. Norgan N. 2000. Long-term physiological and economic consequences Carnethon MR, De Chavez PJD, Biggs ML, Lewis CE, Pankow JS, of growth retardation in children and adolescents. Proc Nutr Soc. – Bertoni AG, Golden SH, Liu K, Mukamal KJ, Campbell-Jenkins B. 59:245 256. 2012. Association of weight status with mortality in adults with inci- Perkins KA. 1992. Effects of tobacco smoking on caloric intake. Br J – dent diabetes. JAMA. 308:581–590. Addict. 87:193 205. Cooper SJ, Van der Hoek GA. 1993. Cocaine: a microstructural analysis Petersson A, Olsson-Mortlock C, Thiblin I, Rajs J, Eksborg S. 2004. of its effects on feeding and associated behaviour in the rat. Brain Nutritional status of deceased illicit drug addicts in Stockholm, – Res. 608:45–51. Sweden: a longitudinal medicolegal study. J Forensic Sci. 49:1 10. Crossin R, Cairney S, Lawrence A, Duncan J. 2016. Adolescent inhalant Pryor GT. 1991. A toluene-induced motor syndrome in rats resembling abuse leads to other drug use and impaired growth; implications for that seen in some human solvent abusers. Neurotoxicol Teratol. – diagnosis. Aust NZ J Public Health. 41:99–104. 13:387 400. Dick A, Simpson A, Qama A, Andrews Z, Lawrence A, Duncan J. 2015. Reed RB, Stuart HC. 1959. Patterns of growth in height and weight – Chronic intermittent toluene inhalation in adolescent rats results in from birth to eighteen years of age. Pediatrics. 24:904 921. metabolic dysfunction with altered glucose homeostasis. Br J Shah R, Vankar G, Upadhyaya HP. 1999. Phenomenology of gasoline Pharmacol. 172:5174–5187. intoxication and withdrawal symptoms among adolescent in India: a Dulloo AG, Jacquet J, Seydoux J, Montani J-P. 2006. The thrifty ‘catch- case series. Am J Addict. 8:254–257. up fat’phenotype: its impact on insulin sensitivity during growth tra- Siegel P, Clopper R, Stabler B. 1990. Psychological impact of signifi- jectories to obesity and metabolic syndrome. Int J Obes Relat Metab cantly short stature. Acta Paediatr Scand Suppl. 377:14–18. Disord. 30:S23–S35. Sumithran P, Proietto J. 2013. The defence of body weight: a physio- Ersche KD, Stochl J, Woodward JM, Fletcher PC. 2013. The skinny on logical basis for weight regain after weight loss. Clin Sci. cocaine: insights into eating behavior and body weight in cocaine- 124:231–241. dependent men. Appetite. 71:75–80. Thornhill J, Hirst M, Gowdey C. 1976. Disruption of diurnal Glaser HH, Massengale ON. 1962. Glue-sniffing in children. Deliberate feeding patterns of rats by heroin. Pharmacol Biochem Behav. inhalation of vaporized plastic cements. JAMA. 181:300–303. 4:129–135. Grebenstein PE, Thompson IE, Rowland NE. 2013. The effects of Williams MT, Moran MS, Vorhees CV. 2004. Behavioral and extended intravenous nicotine administration on body weight and growth effects induced by low dose methamphetamine administra- meal patterns in male Sprague-Dawley rats. Psychopharmacology. tion during the neonatal period in rats. Int J Develop Neurosci. 228:359–366. 22:273–283. Rose Crossin (737900)

Chapter 3 – A systematic review and meta-analysis on the growth impacts of inhalant abuse The aim of this study was quantify the magnitude of the effect on inhalant abuse (in clinical studies) and toluene exposure (in pre-clinical models) on body weight and height using previously published literature. This took into account study characteristics, to identify potential moderators to these effects on body weight and height. The rationale for this aim was firstly to build upon Chapter 2, and to determine whether the findings from the human cohort were consistent with the body of literature. Secondly, to determine the magnitude of these effects on body weight and height, including whether there was a dose-response relationship evident, and third, to determine if effects varied by study characteristics such as sex, species, and age at first exposure. This study would provide beneficial information for the inhalant abuse field more broadly, and also enable future experiments in an animal model to be guided through determination of the factors that would need to be controlled for. This was described in the paper: Growth changes after inhalant abuse and toluene exposure: a systematic review and meta-analysis of human and animal studies.

Paper citation:

Crossin, R., Lawrence, A. J., Andrews, Z. B., Churilov, L., & Duncan, J. R. (2018). Growth changes after inhalant abuse and toluene exposure: A systematic review and meta-analysis of human and animal studies. Human & Experimental Toxicology, 0960327118792064.

39 Article

Human and Experimental Toxicology 1–16 ª The Author(s) 2018 Growth changes after inhalant abuse Article reuse guidelines: sagepub.com/journals-permissions and toluene exposure: A systematic DOI: 10.1177/0960327118792064 review and meta-analysis of human journals.sagepub.com/home/het and animal studies

R Crossin1,2 , AJ Lawrence1,3, ZB Andrews4 , L Churilov3 and JR Duncan1,5

Abstract Inhalant abuse is a significant public health issue, particularly for adolescents, the predominant group of inhalant users. Adolescence is a critical growth period, and inhalant abuse has been associated with growth impair- ments, including reduced body weight and height. However, the extent to which inhalant abuse affects growth remains unquantified, and potential moderators remain unknown. To address this knowledge gap, a systematic review and meta-analysis of clinical human and preclinical animal studies utilizing toluene exposure (the primary solvent in abused products) was conducted. Five-hundred and sixty-nine studies were screened; 31 met inclusion criteria, yielding 64 toluene-control comparisons for body weight and 6 comparisons for height. Toluene exposure was negatively associated with body weight (d ¼0.73) and height (d ¼0.69). Concen- tration of inhaled toluene, but not duration, moderated the effect of toluene exposure on body weight, with more severe impairments at higher concentrations. Differences in effect size for body weight were observed for study characteristic subgroups including sex, age at first exposure, administration route and species. However, these findings should be interpreted cautiously due to low study numbers. Growth impairments, particularly during adolescence, can cause long-term health consequences. These effects on growth are therefore an important clinical outcome for individuals with a history of inhalant abuse.

Keywords Weight, height, volatile solvent abuse, adolescence

Introduction 1 Inhalant abuse is the deliberate inhalation of products Addiction Neuroscience, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Melbourne, containing volatile solvents, in order to achieve an 1 Victoria, Australia altered mental state. Demographic studies into inha- 2Eastern Health Clinical School, Monash University, Clayton, Vic- lant abuse show that usage patterns are similar across toria, Australia sexes2 and overwhelmingly indicate that abuse is 3Florey Institute of Neuroscience and Mental Health, University most commonly associated with adolescence (partic- of Melbourne, Parkville, Melbourne, Victoria, Australia 4Department of Physiology, Monash University, Clayton, Victoria, ularly young adolescence) patterns that are consistent 3–5 Australia worldwide. For example, population-based surveys 5School of Medicine, University of Adelaide, Adelaide, South Aus- indicate that 19.1% of Australian 12- to 13-year olds tralia, Australia self-report experimenting with inhalants.3 While large population-based surveys do not collect data from Corresponding author: R Crossin, Addiction Neuroscience, Florey Institute of Neu- children under the age of 12, and smaller cohort roscience and Mental Health, University of Melbourne, Parkville, studies have identified that children under the age of Melbourne, Victoria 3010, Australia. 6 12 may also misuse inhalants. Email: [email protected] 2 Human and Experimental Toxicology XX(X)

Preadolescence and adolescence is a critical period characteristics. Firstly, the range of products inhaled of growth, with the peak growth spurt for females by inhalant users is varied and opportunistic and occurring between 10 years and 14 years of age, and includes common household items such as glue, thin- a slightly later spurt for males occurring at 12–15 ners and aerosols. However, a common solvent to all years of age.7 Studies indicate that for many inhalant these products is toluene, which is a colourless, water- users, the duration of abuse occurs over periods insoluble, aromatic hydrocarbon. Inhalant users will greater than 10 years, with users reporting abuse pat- preferably inhale pure toluene if it is available,20 and terns equivalent to intermittent high usage,8 thus over- thus toluene exposure is utilized as an exemplar for lapping with the period when rapid growth is inhalant abuse in animal models in the majority of occurring in many young inhalant users.3 It is, there- studies.21 Study design must consider toluene dosage fore, unsurprising that inhalant abuse has an effect on which when the administration route is inhalation will growth, which has been observed in both males6,9 and be reflected as a concentration in parts per million females10; however, this effect is poorly understood. (ppm). Human inhalant users will often inhale via a Case studies from the 1960s report that inhalant solvent-soaked cloth or directly from a bag, and the users present as emaciated,9 and a reduction in weight concentration of inhaled toluene from this method is is well-accepted as a consequence of inhalant abuse, estimated to range from 3000 ppm to 15,000 ppm.21 It to the extent that it is often listed as a warning sign of is important to note that exposure to toluene may also this type of drug abuse.1 Indeed weight impairments occur in the industrial setting; however, this occurs in can be so severe in humans that inhalant abuse is a different pattern to abuse with exposure at lower associated with failure to thrive (FTT).6 Weight concentrations for extended durations. Secondly, impairments (either a loss of weight or reduction in many humans abuse inhalants in a chronic yet inter- weight gain) is also a common outcome in animal mittent nature.8 Thus, a study design must include a experiments that utilize toluene exposure as a model pattern of exposure in minutes/day and days/week for of inhalant abuse, occurring immediately after the a specified period of time. Despite the diverse range exposure period,11–13 and body weight is often of exposure patterns used experimentally, this can be included as a descriptive statistic, even if it was not quantified as a variable called ‘exposure hours’, the key outcome under consideration. However, the which can then be multiplied by the dosage to give effects of toluene exposure on body weight are not ‘total toluene exposure’. The third issue is that of consistent in effect size, suggesting that unknown administration route. Inhalation is the most reflective moderators may play a role in the effect of toluene of human usage but requires specialized equipment exposure on weight. and thus some models utilize intraperitoneal (IP) Given that the adolescent growth spurt is reflected injections or gavage to administer toluene. How the in a change to individuals’ weight and height, it is amount of toluene administered is reported will surprising that the effects of inhalant abuse on height depend on the administration route, with inhalation have not been studied to the same degree as weight. reported as a concentration in ppm, IP injections as Height impairments arising from inhalant abuse have milligrams/kilogram (mg/kg) and gavage as millili- been reported in both humans6,14 and animal models tres/kilogram (mL/kg). The final issue is that of utilizing toluene exposure,15 and in humans this effect experimental animal. Rats and mice are the predomi- has been shown to persist into sustained abstinence.16 nant species used in inhalant abuse models, with rats Again the size of this effect has never been quantified, having equivalent sensitivity with humans to and the potential moderators remain unknown. toluene,22 but choices must still be made about sex Despite our lack of understanding of the factors mod- and the age at which exposure will begin, bearing in erating inhalant-induced changes to growth para- mind the association between inhalant abuse and meters, the effects of inhalant abuse on weight and young adolescence. height are important outcomes. This is especially due To date, the extent to which inhalant abuse affects to the potential for long-term health impacts arising growth patterns have not been quantified, and the from alterations to growth,17–19 even if an individual effects of potential moderators are not well- with a history of abusing inhalants remains abstinent. understood. To address this knowledge gap, we The majority of the research into the physiological conducted a systematic review of human and animal consequences of inhalant abuse is derived from ani- studies and applied meta-analytic techniques to cal- mal models, which incorporate five main study culate an effect size d. The primary aim of the study Crossin et al. 3

Table 1. Key words (in greyed boxes) and synonyms (below) used as alternative terms for the search strategy. Inhalant abuse Adolescence Weight Height Body mass index Volatile solvent abuse Teenage Body weight Length BMI VSA Youth Emaciation Spine length Adiposity Toluene Young adult Malnutrition Stunting Waist circumference Petrol sniffing Overweight Linear growth Z-score Glue sniffing Obesity Weight-to-height ratio Huffing Underweight Cachexia Wasting Failure to thrive was to examine the effects of inhalant abuse (in studies, only those that utilized toluene as the inha- humans) and toluene exposure (in animals) on weight lant were included in this meta-analysis (excluding and height, hypothesizing that ‘as amount of toluene animal models of inhaled benzene or xylene), as this administered (in ppm, mg/kg or mL/kg) increases, directly links the experimental model to the products height and body weight will decrease’. A secondary typically abused by humans (e.g. glue, paint and sol- aim was to explore the effect on height and body vents). Given that growth data are often reported as a weight of a range of study characteristics that are com- descriptive statistic in studies, without being a pri- monly encountered in inhalant abuse studies, pattern of mary outcome, it was not possible to limit the search exposure (including exposure duration and total to keywords such as inhalant þ weight. Therefore, a toluene exposure), administration route and model range of strategies were utilized including keyword (including species, sex and age at first exposure). searches, searches using synonyms for describing growth patterns (Table 1) and keyword searches of Methods physiological outcomes relative to toluene exposure where it might be plausible that body weight would This study was registered with the open access data- be reported (e.g. reproduction, motor syndrome and base PROSPERO (international prospective register food intake). of systematic reviews) at inception (registration num- Each combination of keyword and synonym was ber CRD42017069002; https://www.crd.york.ac.uk/ used as search strings; this strategy meant that 84 prospero/display_record.php?RecordID¼69002) and 23 unique searches (provided as search strings in Online conducted in accordance with PRISMA guidelines. Supplemental Table 1) were performed in PubMed, The completed PRISMA flow chart is provided as which yielded a total of 622 potential studies; 53 of Online Supplemental material. those were duplicates, leaving 569 studies that were checked to determine whether they included a Systematic literature review description of height or weight. Of the 569 studies, Peer-reviewed studies of inhalant abuse and toluene only 52 reported on at least one of these outcome exposure were collated via extensive literature measures. Studies were then excluded if they did not searches, searched for in PubMed. To be included, provide the mean of height or weight along with a studies must have provided a measure of height or measure of variance, for both the treatment and the weight that was taken immediately or shortly after control groups. This criterion reduced the number of exposure, sample sizes in each group along with a eligible studies from 52 to 31, with 21 studies measure of variance such as standard deviation (SD) excluded. It was deemed that this number of studies or standard error of the mean (SEM), in order for an was sufficient to proceed from systematic review to effect size to be calculated. Studies that focused on meta-analysis, and these 31 studies were then used for height or weight effects occurring in sustained absti- the meta-analysis. Figure 1 shows the PRISMA flow nence (i.e. after exposure to toluene had ceased) chart; the 21 excluded studies with reasons box refer were not the focus of this study and were not to the 21 studies discussed above. Risk of bias within included. Only published, peer-reviewed studies the included studies was assessed using the SYRCLE were considered for inclusion. For the animal risk-of-bias tool for animal studies,24 acknowledging 4 Human and Experimental Toxicology XX(X)

Figure 1. /prismastatement/flowdiagram (PRISMA) flow chart showing study numbers. Source: Moher et al.23 For more information, see www.prisma-statement.org. that four of the included studies were clinical obser- statistics computed was Cohen’s d, by comparing the vational studies, however, it was not considered mean body weight and/or height from the toluene group appropriate to utilize multiple risk of bias tools. to the mean body weight and/or height from the control Results of the assessment are provided as Online Sup- group. Then, 95% confidence intervals were computed plemental Table 2. for each d. Heterogeneity is reported using the I2 statis- tic, which describes the percentage of variation across studies that is due to heterogeneity rather than chance. Meta-analysis techniques Following Higgins et al.,50 we consider I2 values of The software program Review Manager (RevMan) ver- 25%,50% and 75% as low, moderate and high, respec- sion 5.3, Copenhagen: The Nordic Cochrane Centre, tively. Note that these categories do not refer to the The Cochrane Collaboration, 2014, was used to analyse absolute amount of observed heterogeneity, but rather the data and conduct the meta-analysis. An effect size to the proportion of the observed effect variance that statistic was computed for each toluene-to-control com- would remain if the sampling error were to be elimi- parison; the effect size was estimated by pooling indi- nated, that is, if we were to be able to observe the true vidual effects using a random-effects model. The effect size for all studies in the analysis. Crossin et al. 5

When entering the data into RevMan, the reportedpffiffiffiffi postnatal day and/or body weight, from which the SEM values were converted to SD (SD ¼ SEM N). category was calculated.53 Pearson’s correlation was As many of the studies utilized multiple treatment used to explore the relationship between concentra- groups, often due to testing more than one concentra- tion and exposure for the identified experimental tion of toluene, a multiple-comparisons correction models. There are limited published data available for was required.51 The approach taken was to compute toluene exposure, and owing to incomplete data in all an ‘effective N’ for the control group that was being categories, a meta-regression could not be underta- utilized for more than one treatment comparison by ken, which would allow the relationship between dividing N by the number of total comparisons. Thus, study characteristics to be explored more fully. The each toluene-to-control comparison was entered indi- small number of studies also did not provide sufficient vidually into RevMan, with 64 unique comparisons power for the inclusion of funnel plots to assess pub- for body weight and 6 unique comparisons for height lication bias. The approach taken was therefore arising from the 31 studies. exploratory in nature, estimating the effects of the The studies identified in the systematic review uti- various study characteristics. All p values for individ- lized a wide range of concentrations of toluene (min- ual comparisons (seven in total) were therefore imum 80 ppm, maximum 10,000 ppm). To analyse reported, without multiplicity correction, and all concentration effectively, these were collapsed into Cohen’s d results are reported with the 95% confi- the following groups: 0–500 ppm, 501–2000 ppm, dence interval in brackets. 2001–5000 ppm and >5000 ppm. These groupings were chosen for consistency with previous meta- analysis on toluene exposure52 and provided an Results approximately even pool of comparisons across the Overall effect sizes and study characteristics subgroups. Additional analysis was performed post A summary of the study characteristics and effect hoc comparing occupational exposure concentrations sizes is provided in Table 2. The overall effect size ( 1000 ppm) with inhalant abuse concentrations on body weight was 0.73 [95% CI 0.99, 0.47], (>1000 ppm). Studies using a non-inhalation route with I2 of 76% categorized as high heterogeneity such as IP injection or gavage were not assigned a (Figure 2), and the overall effect size on height was concentration. Exposure hours reflected the total 0.69 [95% CI 1.16, 0.21], with I2 of 65% cate- duration of exposure to toluene and were calculated gorized as moderate–high heterogeneity (Figure 3), by multiplying the exposure duration in hours and the indicating that exposure to toluene resulted in signif- total number of exposures. For example, 1 h/day and 3 icantly decreased body weight and height, though the days/week for 4 weeks would be reported as 12 expo- relatively high heterogeneity suggests that subgroups sure hours. Total toluene exposure (in ppm h) was may exist that affect the outcome.50 calculated by multiplying exposure hours and the con- centration in ppm, and then collapsed into subgroups of low (0–50,000 ppm h), medium (50,001–200,000 The effect of toluene concentration on body ppm h), high (>200,000 ppm h), which were chosen to weight and height provide approximately even distribution of studies As hypothesized, toluene concentration was a clini- across the subgroups. Fourteen comparisons could not cally meaningful moderator of body weight ( 2 ¼ be allocated a concentration (due to either utilizing a 25.50, df ¼ 3, p < 0.00001), as shown in Figure 4, non-inhalation route or because it was a human study with a full forest plot in Online Supplemental Figure of inhalation and concentration was not measured) 1. Clear separation between the 95% confidence inter- and did not have exposure hours and, therefore, could vals was observed between the upper and lower con- not have total toluene exposure computed. centration subgroups, suggestive of a concentration– Each study characteristic was assigned subgroups response relationship, though there was overlap (e.g. sex: male and female), and 2 tests were used to between the confidence intervals of the two middle quantify the differences between subgroups for each subgroups. The lowest concentration subgroup (0– study characteristic. Characterization of age at first 500 ppm) had no observable effect on body weight, exposure was into either adolescent or adult and was with decreases in body weight observed in the three based on data extracted from studies; some studies upper concentration subgroups. Furthermore, when directly stated adult/adolescent, whereas others stated concentration was differentiated post hoc by 6 Table 2. Summary of study characteristics and effect sizes used in the meta-analysis.a Effect size (d) for body Effect size Toluene amount Study ID weight with [95% CI] (d) for height (ppm unless stated) Exposure hours Total toluene exposure Admin. route Species Sex Age at first admin. Bushnell et al.25 1-1 0.22 [1.11, 1.55] 100 300 Low Inhalation Mouse M Adult 1-2 0.30 [1.04, 1.63] 1000 300 High Inhalation Mouse M Adult 1-3 0.27 [1.61, 1.06] 3000 300 High Inhalation Mouse M Adult Chan et al.26 2-1 0.06 [0.99, 1.10] 500 mg/kg IP injection Rat M Adolescent 2-2 0.07 [1.12, 0.98] 500 mg/kg IP injection Rat F Adolescent 2-3 0.51 [0.56, 1.59] 500 mg/kg IP injection Rat M Adolescent 2-4 0.31 [1.36, 0.75] 500 mg/kg IP injection Rat F Adolescent Chen et al.27 3-1 0.04 [0.73, 0.80] 500 mg/kg IP injection Rat M Adolescent 3-2 0.17 [0.59, 0.93] 500 mg/kg IP injection Rat M Adolescent Chien et al.28 4-1 0.03 [1.08, 1.02] 500 mg/kg IP injection Rat M Adolescent 4-2 0.47 [0.60, 1.54] 500 mg/kg IP injection Rat M Adolescent Crossin et al.6 5-1 0.68 [1.09, 0.26] 0.79 [1.21, 0.37] Not stated Inhalation Human M Adolescent Dick et al.29 6-1 1.34 [2.43, 0.24] 10,000 12 Medium Inhalation Rat M Adolescent 6-2 2.57 [3.38, 1.76] 10,000 12 Medium Inhalation Rat M Adolescent Dick et al.30 7-1 0.17 [0.97, 0.64] 3000 12 Low Inhalation Rat M Adolescent 7-2 0.20 [0.60, 1.00] 3000 24 Medium Inhalation Rat M Adolescent Dick et al.31 8-1 0.40 [1.35, 0.54] 3000 20 Medium Inhalation Rat M Adolescent 8-2 1.95 [2.86, 1.03] 10,000 12 Medium Inhalation Rat M Adolescent Dick et al.11 9-1 1.92 [2.68, 1.16] 10,000 12 Medium Inhalation Rat M Adolescent Duncan et al.12 10-1 0.18 [1.06, 0.70] 3000 12 Low Inhalation Rat M Adolescent 10-2 1.21 [2.18, 0.24] 3000 24 Medium Inhalation Rat M Adolescent Duncan et al.32 11-1 3.14 [4.80, 1.49] 10,000 9 Medium Inhalation Rat M Adolescent Dundaroz et al.33 12-1 0.15 [0.68, 0.38] 0.25 [0.78, 0.28] Not stated Inhalation Human M Adolescent Funada et al.34 13-1 1.33 [0.39, 3.05] 350 1.5 Low Inhalation Mouse M Adult 13-2 0.15 [1.70, 1.40] 700 1.5 Low Inhalation Mouse M Adult 13-3 0.25 [1.31, 1.80] 2500 1.5 Low Inhalation Mouse M Adult 13-4 0.27 [1.29, 1.82] 3200 1.5 Low Inhalation Mouse M Adult Furlong et al.35 14-1 1.84 [3.61, 0.08] 10,000 12 Medium Inhalation Rat M Adolescent Gospe et al.36 15-1 1.46 [2.42, 0.50] 1 mL/kg Gavage Rat F Adolescent Gotohda et al.37 16-1 1.11 [2.12, 0.10] 1500 28 Low Inhalation Rat M Adolescent Ishigami et al.38 17-1 4.58 [9.27, 0.12] 1500 80 Medium Inhalation Rat M Adolescent Jenkins et al.39 18-1 0.70 [0.23, 1.62] 1085 240 High Inhalation Rat M and F Adolescent 18-2 0.78 [0.18, 1.73] 107 2160 High Inhalation Rat M and F Adolescent 18-3 3.79 [5.27, 2.31] 1085 240 High Inhalation Guinea pig M and F Adolescent 18-4 0.60 [1.48, 0.28] 107 2160 High Inhalation Guinea pig M and F Adolescent 18-5 2.92 [5.76, 0.09] 1085 240 High Inhalation Dog M and F Adolescent 18-6 0.57 [2.27, 1.13] 107 2160 High Inhalation Dog M and F Adolescent 18-7 0.22 [1.61, 1.18] 1085 240 High Inhalation Monkey M and F Adolescent 18-8 1.19 [0.38, 2.76] 107 2160 High Inhalation Monkey M and F Adolescent Johnson40 19-1 0.76 [1.70, 0.18] 1000 160 Medium Inhalation Rat M Adolescent Martı´nez-Alfaro et al.41 20-1 3.14 [5.29, 0.98] 3000 21 Medium Inhalation Rat M Adolescent Moron et al.13 21-1 1.59 [2.96, 0.22] 1.3 mL/kg IP injection Rat M Adult Olgar et al.14 22-1 1.70 [2.16, 1.23] 1.46 [1.91, 1.01] 7.63 + 5.69 tubes (50 mL/tube) Inhalation Human M Adolescent Ono et al.42 23-1 2.87 [5.31, 0.43] 4000 70 High Inhalation Rat M Adolescent 23-2 4.09 [7.25, 0.93] 6000 70 High Inhalation Rat M Adolescent Pryor15 24-1 2.70 [3.86, 1.55] 2000 616 High Inhalation Rat M Adolescent 24-2 4.91 [7.18, 2.65] 0.11 [1.24, 1.02] 2200 1288 High Inhalation Rat M Adolescent (continued) Table 2. (continued)

Effect size (d) for body Effect size Toluene amount Study ID weight with [95% CI] (d) for height (ppm unless stated) Exposure hours Total toluene exposure Admin. route Species Sex Age at first admin. 24-3 5.46 [7.91, 3.00] 0.31 [1.45, 0.83] 4400 644 High Inhalation Rat M Adolescent 24-4 6.00 [8.65, 3.35] 0.66 [1.82, 0.50] 6200 322 High Inhalation Rat M Adolescent Roberts et al.43 25-1 0.22 [1.13, 0.69] 100 570 Medium Inhalation Rat F Adolescent 25-2 0.43 [1.35, 0.50] 500 570 High Inhalation Rat F Adolescent 25-3 0.66 [1.63, 0.31] 2000 570 High Inhalation Rat F Adolescent 25-4 0.23 [1.14, 0.69] 2000 570 High Inhalation Rat F Adolescent 25-5 1.18 [2.19, 0.17] 2000 570 High Inhalation Rat F Adolescent Roberts et al.44 26-1 0.16 [0.75, 1.06] 250 54 Low Inhalation Rat F Adult 26-2 0.05 [0.87, 0.96] 750 54 Low Inhalation Rat F Adult 26-3 0.17 [0.75, 1.09] 1500 54 Medium Inhalation Rat F Adult 26-4 1.09 [2.05, 0.14] 3000 54 Medium Inhalation Rat F Adult Saillenfait et al.45 27-1 0.50 [0.37, 1.37] 500 84 Low Inhalation Rat F Adolescent 27-2 0.43 [1.29, 0.42] 1500 84 Medium Inhalation Rat F Adolescent Schiffer et al.46 28-1 1.88 [3.39, 0.37] 5000 3 Low Inhalation Rat M Adolescent Uchino et al.47 29-1 0.84 [1.32, 0.36] Not stated Inhalation Human F Adolescent von Euler et al.48 30-1 0.24 [0.75, 0.27] 80 120 Low Inhalation Rat M Adolescent Yoon et al.49 31-1 0.25 [0.38, 0.87] 7000 4 Low Inhalation Rat M Adolescent

IP: intraperitoneal. aThirty-one studies were included in the meta-analysis, yielding 64 unique toluene-to-control comparisons for body weight and 6 unique comparisons for height. ID is the study number followed by the comparison number. The characteristics of each toluene-to-control comparison are outlined in Table 2, along with the mean effect size and 95% confidence interval in brackets. Boxes are greyed out when there are no data available. Total toluene exposure was first calculated by multiplying concentration and exposure hours (ppm h). These were then collapsed into categories based on low (0–50,000 ppm h), medium (50,001–200,000 ppm h), high (>200,000 ppm h). 7 8 Human and Experimental Toxicology XX(X)

Figure 2. Exposure to toluene impaired body weight. Bars represent the effect size for each study with a 95% confidence interval. Multiple toluene-to-control comparisons (c) existed within a single study, and these are noted as c1, c2, and so on. occupational-level exposure (1000 ppm) or inhalant p < 0.00001). Only inhalant abuse exposure had an abuse exposure (>1000 ppm), this was also a signifi- effect on body weight (Online Supplemental Figure cant moderator of body weight ( 2 ¼ 25.81, df ¼ 1, 2), with no observable effect on body weight for Crossin et al. 9

Figure 3. Exposure to toluene impaired height. Bars represent the effect size for each study with a 95% confidence interval. Multiple toluene-to-control comparisons (c) existed within a single study, and these are noted as c1, c2, and so on.

intervals were observed. However, there was a sig- nificant negative relationship (Pearson’s correlation coefficient ¼0.366, p ¼ 0.009) between concen- tration and exposure hours in the experimental mod- els within the studies, and the highest concentrations only occurred in the lowest exposure subgroup (Figure 6). Total toluene exposure was computed to account for this relationship, and increased total toluene exposure was associated with decreased body weight ( 2 ¼ 21.55, df ¼ 2, p < 0.00001, Table 3, Figure 5 and Online Supplemental Figure Figure 4. As toluene concentration increased, body 4), with the low exposure subgroup having no very weight was more impaired. The mean effect size for each small effect on body weight; however, there was subgroup within toluene concentration is shown with a 95% confidence interval. Only studies that cited a specific overlap between the confidence intervals of the concentration of toluene (in ppm) were included, which medium and high subgroups, both of which showed excluded 14 comparisons. Inhaled concentration was a large effect sizes. significant moderator of body weight, with the highest Administration route affected body weight ( 2 ¼ negative effect size observed at the highest concentration. 18.42, df ¼ 2, p ¼ 0.0001, Table 3, Figure 5 and Online Supplemental Figure 5), with toluene adminis- occupational-level exposure. The 14 comparisons that tered via IP injection having no effect on body weight, could not be ascribed a concentration in ppm were not but decreases being observed for both inhalation and able to be included in these subgroup analyses. In gavage, with large effect sizes. Differences in the addition, due to the unavailability of data in all cate- effect of toluene exposure on body weight were also gories for height, these analyses could not be com- observed for both species ( 2 ¼ 14.95, df ¼ 5, p ¼ pleted for height as an outcome. 0.01, Table 3, Figure 5 and Online Supplemental Fig- ure 6) and sex ( 2 ¼ 5.32, df ¼ 1, p ¼ 0.02, Table 3, Figure 5 and Online Supplemental Figure 7). Body Estimating the effect of study characteristics on weight decreases, with large effect sizes, were body weight observed for humans and rats following toluene expo- The effect of all study characteristics on body weight sure, but results were inconclusive for dogs and gui- was investigated, and the findings are summarized in nea pigs exacerbated by the low number of studies for Table 3 and Figure 5 (full forest plots in Online Sup- comparison, and results for monkeys and mice plemental Figures 3 to 8). trended towards an increased body weight following There was not a clear relationship between expo- toluene exposure, despite all these species undergoing sure hours and body weight ( 2 ¼ 0.65, df ¼ 3, p ¼ exposure via inhalation. While both males and 0.89, Table 3, Figure 5 and Online Supplemental females showed a decrease in body weight following Figure 3), and overlaps between the confidence toluene exposure, the effect size was greater for males 10 Human and Experimental Toxicology XX(X)

Table 3. The effects of toluene exposure on body weight, by subgroup. Moderator kI2 (%) Effect size (d) [95% confidence interval] p Value for subgroup differences Overall 64 76 0.73 [0.99, 0.47] N/A Inhaled concentration 0–500 ppm 11 21 0.05 [0.28, 0.38] <0.00001 501–2,000 ppm 16 72 0.78 [1.33, 0.23] 2001–5000 ppm 14 75 1.12 [1.79, 0.45] >5000 ppm 9 86 2.18 [3.18, 1.18] Exposure hours 0–20 15 80 0.91 [1.53, 0.29] 0.89 21–100 13 66 0.66 [1.21, 0.11] 101–500 10 82 0.94 [1.82, 0.06] >500 12 81 0.98 [1.74, 0.22] Total toluene exposure Low 14 19 0.06 [0.35, 0.23] <0.00001 Medium 16 74 1.22 [1.75, 0.70] High 20 83 1.36 [2.09, 0.64] Administration route Inhalation 54 78 0.85 [1.14, 0.55] 0.0001 IP injection 9 0 0.01 [0.32, 0.33] Gavage 1 N/A 1.46 [2.42, 0.50] Species Human 4 85 0.85 [1.45, 0.24] 0.01 Rat 47 78 0.83 [1.15, 0.51] Mouse 7 0 0.23 [0.33, 0.78] Guinea pig 2 92 2.14 [5.26, 0.99] Dog 2 49 1.46 [3.70, 0.78] Monkey 2 42 0.44 [0.94, 1.81] Sex Male 41 80 0.95 [1.30, 0.59] 0.02 Female 15 33 0.42 [0.70, 0.14] Age at first exposure Adolescent 52 79 0.88 [1.17, 0.59] 0.001 Adult 12 16 0.09 [0.48, 0.29]

IP: intraperitoneal; k ¼ the number of effect sizes (i.e. the number of toluene-to-control comparisons); I2 ¼ heterogeneity.

than females. Age at first exposure also had an effect (2001–5000 ppm and >5000 ppm) and exposure hours on body weight ( 2 ¼ 10.28, df ¼ 1, p ¼ 0.001, Table (101–500 h and >500 h). These results are not shown, 3, Figure 5 and Online Supplemental Figure 8), with due to the incompleteness of the data set. adolescents showing decreased body weight follow- ing toluene exposure, whereas there was no clear effect on body weight for adults, and the confidence Discussion interval crossed zero. We conducted a systematic review and meta- analysis of the inhalant abuse literature, incorporat- ing the use of toluene exposure as an experimental Estimating the effect of study characteristics on model of inhalant abuse and including both human height and animal studies. We demonstrate that inhalant Because of the small number of comparisons for abuse in humans and toluene exposure in animal height, full analysis of all study characteristics could models are associated with significant impairments not be undertaken. No characteristic had all subgroups to both body weight and height, with effect sizes that represented, and the only characteristics for which would be categorized as medium/large according to there was more than one subgroup represented for Cohen.54 Furthermore, the results indicate a concen- height were species (human and rat), concentration tration–response relationship between inhaled Crossin et al. 11

growth have the potential to affect an individual’s long-term health outcomes, through both physiolo- gical and psychological consequences.17–19 The finding that toluene concentration is a clini- cally meaningful moderator of the effect of toluene on body weight is juxtaposed by the non-significant effect of exposure hours, with similar medium–large effect sizes observed across the four subgroups. This may be due to the experimental design, where exposure to the highest concentrations have shorter exposure hours, thus this result may be confounded by the uneven distribution of concentrations within those experimental models. We attempted to over- come this issue by computing total toluene exposure, as a way of combining both toluene concentration and the duration of exposure. Total toluene exposure affected body weight more significantly than expo- sure hours (time) alone, a finding consistent with Bowen et al.55; however, there was no expected dis- tinction between the medium and the high exposure Figure 5. The effect size of toluene exposure on body weight differed by study characteristics, such as exposure subgroups. This suggests that while both toluene con- method, species and age at first exposure. The effect of centration and time in combination (i.e. total toluene each study characteristic on body weight was investigated exposure) affect body weight, the relative importance and the mean effect size for each subgroup within the study of these two factors requires further exploration. characteristic is shown with a 95% confidence interval. For The additional study characteristics of administra- exposure hours and total toluene exposure, only studies tion route, species, sex and age at first exposure meet that cited a specific concentration of toluene (in ppm) were the threshold for significance with p values less than included, which excluded 14 comparisons. 0.05; however, these results should be interpreted with caution. Some findings are consistent with the literature; for example, toluene exposure has been previously associated with weight gain in mice.52 It would also be expected that toluene would have a larger effect during adolescence, when growth is accelerating, as opposed to adulthood when growth (particularly height) is more stable. However, data are not consistently available across all subgroups. For example, in the systematic review, the studies with mice utilized only adult animals, whereas the guinea pig, dog and monkey data all came from adolescent animals. Therefore, we cannot identify whether it is Figure 6. There is an inverse relationship between species, age at first exposure or both that is affecting ¼ ¼ exposure hours and concentration (r 0.3657 p 0.009, the body weight. Furthermore, having identified a Pearson’s correlation test) within the experimental models concentration–response relationship, we found dif- used for the identified studies, demonstrating that the highest concentrations of toluene have the shortest ferences in the concentrations of toluene that differ- exposure hours, and vice versa. This analysis excluded the ent subgroups within each study characteristic were 14 comparisons that did not report a toluene concentra- exposed to. For example, in the highest inhaled con- tion (in ppm). centration subgroup of >5000 ppm, there are only males and no females, only adolescents and no toluene concentration and body weight impairment, adults and only rats and no other animal species. with low concentrations (<500 ppm) having no sig- Given that higher effect sizes were observed in nificant effect on body weight. These changes to males, adolescents and rats, it is possible that this 12 Human and Experimental Toxicology XX(X) is simply reflecting the response to the high concen- individual than to a developing foetus. In contrast to tration of toluene, rather than sex, age and species our findings, Callan et al. did not identify a linear moderating the relationship between toluene expo- concentration–response relationship and observed a sure and growth changes. reversal of the weight impairment trend at the highest It is important that valid animal models of toluene concentration of inhaled toluene but did find a linear exposure exist, which reflect human patterns of inha- relationship between total toluene exposure and lant abuse and which yield comparable outcomes. The weight impairments.52 This may reflect the fact that, differences between effect sizes for administration due to the distribution of the available data, the same route are of concern, particularly as toluene given via subgroups for total toluene exposure could not be IP injection resulted in no significant effect on body used between the two meta-analyses. It is also possi- weight. The data for gavage is difficult to interpret ble that for the exposed individual, toluene concen- due to the low number of studies, but exposure via tration is a more meaningful variable, whereas for a inhalation appears to yield results more consistent developing foetus, it is the total toluene exposure that with clinical outcomes. Rats are the most common causes the greatest effect on birth weight. Consistent animal model used for inhalant abuse, and it is posi- with our findings, Callan et al. found that toluene tive to note the large overlap in the confidence inter- exposure resulted in increased birth weight for mice, vals between rats and humans, suggesting that they compared to rabbits and rats, suggesting that mice are appear to be a valid experimental model. However, as not a good experimental model for exploring the noted, at the highest concentrations, only male rats are growth effects of inhalant abuse.52 represented. Given that inhalant abuse has similar To understand the relationship between inhalant prevalence across sexes,56 future animal experiments abuse and growth more fully, there are a range of need to consider whether the concentration–response experiments that would assist in filling the identified relationship between male and female rats is equiva- knowledge gaps, and while studies may measure lent. This is particularly relevant as only one human weight, weight data are not always included in the inhalant abuse study could be found that directly com- publication but is a valuable output to report on. pared the growth effects of inhalant abuse between Firstly, experiments should be conducted to under- males and females and showed that weight impair- stand the relative importance of concentration versus ments in females were more severe than in males.47 exposure time, within total toluene exposure. Sec- The lack of studies using females is particularly con- ondly, experiments should be done to understand cerning because a weight impairment and any associ- whether sex, age at first exposure and species do mod- ated loss of adiposity11 during adolescence can cause erate the effect of toluene exposure on body weight, disruptions to pubertal and metabolic processes due to by conducting experiments in which all subgroups the endocrinological activity of adipose tissue.57 have equivalent total toluene exposure. Thirdly, it Furthermore, as shown by Callan et al., exposure to would be beneficial for future experiments to also toluene during pregnancy can impair developmental consider height as a growth outcome, which can be body weight,52 and inhalant abuse is most prevalent in easily measured in experimental animals (crown-to- females during childbearing years.2,3 Thus, inhalant rump length) but is often missed as a descriptive out- abuse in females has the potential to affect not only come. This would allow better understanding of how the individual but also her offspring. toluene exposure affects height and what moderates The effects of inhalant abuse in females have been this effect. explored before in a meta-analysis that considered Weight impairment and emaciation can result in a whether toluene exposure during the prenatal period diverse range of harmful outcomes including affected birth weight of the offspring.52 This meta- impaired cognition, renal failure and osteoporosis.17 analysis only included animal studies, and the only Weight impairment may normalize to re-establish growth outcome was birth weight; however, many homeostasis if the factor suppressing weight is of the study characteristics explored were the same. removed.58 However, this process is associated with The average effect size for weight in the individual rapid weight regain (predominantly through increased from this meta-analysis was 0.73, compared to fat deposition), which then increases the risk of sub- 0.39 for the effect on birth weight following prena- sequent health issues such as visceral adiposity and tal exposure.52 This suggests that the growth impacts insulin resistance.18 Thus, weight impairments can of inhalant abuse may be more severe in the exposed cause long-term health impacts to individuals and are Crossin et al. 13 therefore an important clinical outcome for individu- resistance. This study does not provide mechanistic als with a history of inhalant abuse. Future studies explanation for the effect of toluene on body weight should focus on adolescence as the predominant time and height, which means that we are unable to com- for inhalant use,2,3 overlapping with a critical growth ment on whether the effect on height and weight are period, and seek to incorporate subsequent outcomes common correlated outcomes of the same mechanism such as adiposity and insulin resistance, as well as the or distinct outcomes arising from different mechan- persistence of weight and height effects in abstinence, isms. However, understanding the mechanisms to enable a better understanding of the long-term behind these effects on body weight and height, health consequences of inhalant abuse. including the impact of changes to food intake fol- It should be noted that one limitation of this lowing toluene exposure11,13 is necessary and will meta-analysis is the low number of comparisons, provide information that is relevant to the treatment particularly for the height outcome, and the uneven of those with a history of inhalant abuse. distribution of comparisons across the various sub- In conclusion, this systematic review and meta- groups, which makes interpreting the effects of study analysis forms a comprehensive summary of the characteristics difficult. As we were unaware at the human and animal research literature on the growth time of PROSPERO registration whether there would effects of inhalant abuse and toluene exposure (used be sufficient studies to conduct a meta-analysis and/or as a model of inhalant abuse). We have shown a meta-regression, the methodology was not prospec- medium–large negative effect of toluene exposure tively registered, which is a further limitation of this on body weight, which confirms weight impairment study. Overall, the risk of bias was difficult to assess, as a warning sign of inhalant abuse. Indeed the effect with unclear results for the majority of studies within on body weight is such that it has been previously the performance domain, predominantly attributable associated with FTT in humans, and furthermore, to lack of detailed methods on allocation procedures, identification of FTT can be a strong diagnostic mar- housing protocols and blinding. Due to lack of pre- ker for inhalant abuse in relevant clinical popula- registered protocols for any of the included studies, tions.6 We also show that toluene exposure has a there is a risk of bias due to selective outcome report- medium–large negative impact on height, and despite ing; however, for the majority of the studies, body limited studies, the effect size is similar to that of weight and height were not the primary outcomes of body weight. It is therefore recommended that height investigation; therefore, this may not be a major lim- impairment be added to the warning signs of inhalant itation for this study. We also acknowledge the diffi- abuse and as a potential deterrent to adolescents, for culties inherent in comparing animal studies to human whom height impairment compared to their peers may studies, particularly due to the range of substances be an undesirable outcome. We show that rats and (e.g. glue, paint, petrol, thinners, etc.) that humans exposure by inhalation show results consistent with may inhale. Although all these products contain human findings, but other species and administration toluene, there may be unknown substance-specific routes should be utilized with caution, until future effects. Defining the level of exposure is difficult, experimental research has tested whether these study given the multiple combinations between the amount characteristics do moderate the effect of toluene on of toluene exposure, the duration of that exposure and growth. The effect of sex is also under-studied, mak- how long it occurred for. Although we attempted to ing it difficult to extrapolate these findings to females address these issues through computation of exposure in the clinical setting. Additional outcomes that arise duration and total toluene exposure, this approach from growth changes, for example, insulin resistance does not take into account peak blood concentrations and adiposity should be included in future studies. of toluene (data that would be valuable but is not cited Thus, knowledge gaps need to be filled in order to in these studies), and thus a detailed analysis of the understand potential long-term health risks, stemming pharmacokinetics of toluene under the variety of from growth impairments, for individuals with a his- exposure models could not be undertaken. A further tory of inhalant abuse. limitation of this study is tlack of long-term health impact data arising from the changes to growth, which Author contributions means that while we can quantify the effect size on The study was conceived by RC, AJL, ZBA and JRD. The weight and height, we are unable to relate that to any systematic review was undertaken by RC. RC and LC long-term health outcome such as adiposity or insulin designed the meta-analysis, which was conducted by RC 14 Human and Experimental Toxicology XX(X) and checked by LC. All authors were involved in reviewing velocity, and stages of puberty. Arch Dis Child 1976; the manuscript drafts and approved the final version. 51: 170–179. 8. Lubman D, Yu¨cel M and Lawrence A. Inhalant abuse Declaration of Conflicting Interests among adolescents: neurobiological considerations. Br The authors declared no potential conflicts of interest with J Pharmacol 2008; 154: 316–26. respect to the research, authorship, and/or publication of 9. Glaser HH and Massengale ON. Glue-sniffing in chil- this article. dren: deliberate inhalation of vaporized plastic cements. JAMA 1962; 181: 300–303. Funding 10. Malm G and Lying-Tunell U. Cerebellar dysfunction The authors disclosed receipt of following financial support related to toluene sniffing. Acta Neurol Scand 1980; for the research, authorship and/or publication of this arti- 62: 188–190. cle: This research was supported by the Australian National 11. Dick A, Simpson A, Qama A, et al. Chronic intermit- Health and Medical Research Council (NHMRC) tent toluene inhalation in adolescent rats results in (940835), of which AJL is a Principal Research Fellow metabolic dysfunction with altered glucose homeosta- (1116930) and ZBA is a Career Development Fellow sis. Br J Pharmacol 2015; 172: 5174–5187. (1084344) and the Victorian Government’s Operational 12. Duncan JR, Dick ALW, Egan G, et al. Adolescent Infrastructure Support Scheme. RC is funded by a federal toluene inhalation in rats affects white matter matura- RTP scholarship. Funding bodies had no involvement in tion with the potential for recovery following absti- the design, analysis and decision to publish. nence. PloS One 2012; 7: e44790. 13. Moro´n L, Pascual J, Portillo P, et al. Toluene alters appe- ORCID iD tite, NPY, and galanin immunostaining in the rat R Crossin http://orcid.org/0000-0003-1814-1330 hypothalamus. Neurotoxicol Teratol 2004; 26: 195–200. ZB Andrews http://orcid.org/0000-0002-9097-7944 14. Olgar S, Oktem F, Dindar A, et al. Volatile solvent abuse caused glomerulopathy and tubulopathy in street Supplemental material children. Hum Exp Toxicol 2008; 27: 477–483. Supplemental material for this article is available online. 15. Pryor GT. A toluene-induced motor syndrome in rats resembling that seen in some human solvent abusers. References Neurotoxicol Teratol 1991; 13: 387–400. 1. Alcohol and Other Drugs Council of Australia. Policy 16. Crossin R, Cairney S, John Lawrence A, et al. The position – inhalants. Canberra: Alcohol and other persistence of growth impairments associated with Drugs Council of Australia, 2010. adolescent inhalant abuse following sustained absti- 2. Australian Institute of Health and Welfare. National nence. Addict Res Theory 2017; 26: 183–186. drug strategy household survey 2016. Canberra: 17. Mehler PS and Brown C. Anorexia nervosa–medical Australian Institute of Health and Welfare, 2016. complications. J Eat Disord 2015; 3: 11. 3. Cancer Council Victoria. Australian secondary school 18. Dulloo AG, Jacquet J, Seydoux J, et al. The thrifty students’ use of tobacco, alcohol, and over-the-counter ‘catch-up fat’ phenotype: its impact on insulin sensi- and illicit substances in 2011. Melbourne: Cancer tivity during growth trajectories to obesity and meta- Council of Victoria for Australian Government bolic syndrome. Int J Obes (Lond) 2006; 30: S23–S35. Department for Health and Ageing, 2012. 19. Molinari E, Sartorio A, Ceccarelli A, et al. Psycholo- 4. Nguyen J, O’Brien C and Schapp S. Adolescent inha- gical and emotional development, intellectual capabil- lant use prevention, assessment, and treatment: a liter- ities, and body image in short normal children. ature synthesis. Int J Drug Policy 2016; 31: 15–24. J Endocrinol Invest 2002; 25: 321–328. 5. Dell CA, Gust SW and MacLean S. Global issues in 20. Villatoro JA, Cruz SL, Ortiz A, et al. Volatile sub- volatile substance misuse. Subst Use Misuse 2011; 46: stance misuse in Mexico: correlates and trends. Subst 1–7. Use Misuse 2011; 46: 40–45. 6. Crossin R, Cairney S, Lawrence AJ, et al. Adolescent 21. Bowen SE, Batis JC, Paez-Martinez N, et al. The last inhalant abuse leads to other drug use and impaired decade of solvent research in animal models of abuse: growth; implications for diagnosis. Aust N Z J Public mechanistic and behavioral studies. Neurotoxicol Ter- Health. 2016; 41: 99–104. atol 2006; 28: 636–647. 7. Tanner J and Whitehouse R. Clinical longitudinal stan- 22. Benignus VA, Boyes WK, Kenyon EM, et al. Quanti- dards for height, weight, height velocity, weight tative comparisons of the acute neurotoxicity of Crossin et al. 15

toluene in rats and humans. Toxicol Sci 2007; 100: 35. Furlong TM, Duncan JR, Corbit LH, et al. Toluene 146–155. inhalation in adolescent rats reduces flexible behaviour 23. Moher D, Liberati A, Tetzlaff J, et al. Preferred report- in adulthood and alters glutamatergic and GABAergic ing items for systematic reviews and meta-analyses: signalling. J Neurochem 2016; 139: 806–822. the PRISMA statement. PLoS Med 2009; 6: e1000097. 36. Gospe SM, Saeed DB, Zhou SS, et al. The effects of 24. Hooijmans CR, Rovers MM, de Vries RB, et al. SYR- high-dose toluene on embryonic development in the CLE’s risk of bias tool for animal studies. BMC Med rat. Pediatr Res 1994; 36: 811–815. Res Methodol 2014; 14: 43. 37. Gotohda T, Tokunaga I and Kubo S. Toluene 25. Bushnell PJ, Evans HL and Palmes E. Effects of inhalation-induced adrenocortical hypertrophy and toluene inhalation on carbon dioxide production and endocrinological changes in rat. Life Sci 2005; 76: locomotor activity in mice. Fundam Appl Toxicol 1929–1937. 1985; 5: 971–977. 38. Ishigami A, Tokunaga I, Kubo S, et al. Immunohisto- 26. Chan MH, Tang YC, Chien TH, et al. Toluene expo- chemical study of rat spermatogenesis after toluene-in- sure during the brain growth spurt reduced behavioral halation. Leg Med 2005; 7: 42–46. responses to nicotine in young adult rats: a potential 39. Jenkins L, Jones R and Siegel J. Long-term inhalation role for nicotinic acetylcholine receptors in fetal sol- screening studies of benzene, toluene, o-xylene, and vent syndrome. Toxicol Sci 2008; 101: 286–293. cumene on experimental animals. Toxicol Appl Phar- 27. Chen HH, Lin YR and Chan MH. Toluene exposure macol 1970; 16: 818–823. during brain growth spurt and adolescence produces 40. Johnson AC. Auditory sensitivity in rats exposed to differential effects on N-methyl-D-aspartate receptor- toluene and/or acetyl salicylic acid. Neuroreport mediated currents in rat hippocampus. Toxicol Lett 1992; 3: 1141–1144. 2011; 205: 336–340. 41. Martı´nez-Alfaro M, Ca´rabez-Trejo A, Gallegos- 28. Chien TH, Chan MH, Tang YC, et al. Toluene expo- Corona MA, et al. Thinner inhalation effects on sure during the brain growth spurt reduces behavioral oxidative stress and DNA repair in a rat model of responses to noncompetitive N-methyl-D-aspartate abuse. J Appl Toxicol 2010; 30: 226–232. receptor antagonists in adult rats. Psychopharmacol- 42. Ono A, Kawashima K, Sekita K, et al. Toluene inhala- ogy (Berl) 2005; 182: 468–474. tion induced epididymal sperm dysfunction in rats. 29. Dick AL, Axelsson M, Lawrence AJ, et al. Specific Toxicology 1999; 139: 193–205. impairments in instrumental learning following 43. Roberts L, Bevans A and Schreiner C. Developmental chronic intermittent toluene inhalation in adolescent and reproductive toxicity evaluation of toluene vapor rats. Psychopharmacology (Berl) 2014; 231: in the rat: I. Reproductive toxicity. Reprod Toxicol 1531–1542. 2003; 17: 649–658. 30. Dick AL, Lawrence AJ and Duncan JR. Chronic inter- 44. Roberts L, Nicolich M and Schreiner C. Developmen- mittent toluene inhalation initiated during adolescence tal and reproductive toxicity evaluation of toluene in rats does not alter voluntary consumption of ethanol vapor in the rat: II. Developmental toxicity. Reprod in adulthood. Alcohol 2014; 48: 561–569. Toxicol 2007; 23: 521–531. 31. Dick ALW, Pooters T, Gibbs S, et al. NMDA receptor 45. Saillenfait AM, Gallissot F, Sabate´JP, et al. Develop- binding is reduced within mesocorticolimbic regions mental toxic effects of ethylbenzene or toluene alone following chronic inhalation of toluene in adolescent and in combination with butyl acetate in rats after rats. Brain Res 2015; 1624: 239–252. inhalation exposure. J Appl Toxicol 2007; 27: 32–42. 32. Duncan JR, Gibbs SJ and Lawrence AJ. Chronic inter- 46. Schiffer WK, Lee DE, Alexoff DL, et al. Metabolic mittent toluene inhalation in adolescent rats alters correlates of toluene abuse: decline and recovery of behavioural responses to and MK801. function in adolescent animals. Psychopharmacology Eur Neuropsychopharmacol 2014; 24: 480–486. 2006; 186: 159–167. 33. Dundaroz M, Sarici S, Turkbay T, et al. Evaluation of 47. Uchino A, Kato A, Yuzuriha T, et al. Comparison bone mineral density in chronic glue sniffers. Turk J between patient characteristics and cranial MR find- Pediatr 2002; 44: 326–329. ings in chronic thinner intoxication. Euro Radiol 2002; 34. Funada M, Sato M, Makino Y, et al. Evaluation of 12: 1338–1341. rewarding effect of toluene by the conditioned place 48. von Euler M, Pham TM, Hillefors M, et al. Inhalation preference procedure in mice. Brain Res Protoc 2002; of low concentrations of toluene induces persistent 10: 47–54. effects on a learning retention task, beam-walk 16 Human and Experimental Toxicology XX(X)

performance, and cerebrocortical size in the rat. Exp 54. Cohen J. Statistical power analysis for the behavioral Neurol 2000; 163: 1–8. sciences. 2nd ed. Hilsdale, NJ: Lawrence Earlbaum 49. Yoon JH, Seo HS, Lee J, et al. Acute high-level toluene Associates, 1988. exposure decreases hippocampal neurogenesis in rats. 55. Bowen SE, Batis JC, Mohammadi MH, et al. Abuse Toxicol Ind Health 2016; 32: 1910–1920. pattern of gestational toluene exposure and early post- 50. Higgins JP, Thompson SG, Deeks JJ, et al. Measuring natal development in rats. Neurotoxicol Teratol 2005; inconsistency in meta-analyses. BMJ 2003; 327: 27: 105–116. 557–560. 56. Australian Institute of Health and Welfare. 2010 51. Vesterinen H, Sena E, Egan K, et al. Meta-analysis of National drug strategy household survey report. Can- data from animal studies: a practical guide. J Neurosci berra: Australian Institute of Health and Welfare, Methods 2014; 221: 92–102. 2011. 52. Callan S, Kott J, Cleary J, et al. Changes in develop- 57. Siervogel RM, Demerath EW, Schubert C, et al. mental body weight as a function of toluene exposure: Puberty and body composition. Horm Res 2003; a meta-analysis of animal studies. Hum Exp Toxicol 60: 36–45. 2016; 35: 341–352. 58. Sumithran P and Proietto J. The defence of body 53. Sengupta P. The laboratory rat: relating its age with weight: a physiological basis for weight regain after human’s. Int J Prev Med 2013; 4: 624–630. weight loss. Clin Sci (Lond) 2013; 124: 231–241. The effects of inhalant abuse on growth: a meta-analysis

Supplementary Figure 1 As toluene concentration increases, weight decreases. Bars represent the effect size for each study with a 95% confidence interval. Where multiple toluene-to-control comparisons (c) existed within a single study, these are noted as c1, c2, etc. The effects of inhalant abuse on growth: a meta-analysis

Supplementary Figure 2 When toluene concentration is differentiated between occupational level exposure and inhalant abuse exposure, weight decreases only for inhalant abuse exposure. Bars represent the effect size for each study with a 95% confidence interval. Where multiple toluene-to-control comparisons (c) existed within a single study, these are noted as c1, c2, etc.

The effects of inhalant abuse on growth: a meta-analysis

Supplementary Figure 3 The duration of toluene exposure does not affect body weight. Bars represent the effect size for each study with a 95% confidence interval. Where multiple toluene-to-control comparisons (c) existed within a single study, these are noted as c1, c2, etc. The effects of inhalant abuse on growth: a meta-analysis

Supplementary Figure 4 The effect of total toluene exposure on body weight. Bars represent the effect size for each study with a 95% confidence interval. Where multiple toluene-to- control comparisons (c) existed within a single study, these are noted as c1, c2, etc.

The effects of inhalant abuse on growth: a meta-analysis

Supplementary Figure 5 The route of toluene exposure alters the effect on body weight. Bars represent the effect size for each study with a 95% confidence interval. Where multiple The effects of inhalant abuse on growth: a meta-analysis toluene-to-control comparisons (c) existed within a single study, these are noted as c1, c2, etc.

The effects of inhalant abuse on growth: a meta-analysis

The effects of inhalant abuse on growth: a meta-analysis

Supplementary Figure 6 Species alters the effect of toluene on body weight. Bars represent the effect size for each study with a 95% confidence interval. Where multiple toluene-to- control comparisons (c) existed within a single study, these are noted as c1, c2, etc.

The effects of inhalant abuse on growth: a meta-analysis

Supplementary Figure 7 Toluene exposure decreases body weight for both sexes, but the effect size is different for each sex. Bars represent the effect size for each study with a 95% confidence interval. Where multiple toluene-to-control comparisons (c) existed within a single study, these are noted as c1, c2, etc.

The effects of inhalant abuse on growth: a meta-analysis

Supplementary Figure 8 Toluene exposure impairs body weight more for adolescents than for adults. Bars represent the effect size for each study with a 95% confidence interval. The effects of inhalant abuse on growth: a meta-analysis

Where multiple toluene-to-control comparisons (c) existed within a single study, these are noted as c1, c2, etc.

PRISMA 2009 Checklist

Reported Section/topic # Checklist item on page # TITLE Title 1 Identify the report as a systematic review, meta-analysis, or both. ABSTRACT Structured summary 2 Provide a structured summary including, as applicable: background; objectives; data sources; study eligibility criteria, participants, and interventions; study appraisal and synthesis methods; results; limitations; conclusions and implications of key findings; systematic review registration number. INTRODUCTION Rationale 3 Describe the rationale for the review in the context of what is already known. Objectives 4 Provide an explicit statement of questions being addressed with reference to participants, interventions, comparisons, outcomes, and study design (PICOS). METHODS Protocol and registration 5 Indicate if a review protocol exists, if and where it can be accessed (e.g., Web address), and, if available, provide registration information including registration number. Eligibility criteria 6 Specify study characteristics (e.g., PICOS, length of follow-up) and report characteristics (e.g., years considered, language, publication status) used as criteria for eligibility, giving rationale. Information sources 7 Describe all information sources (e.g., databases with dates of coverage, contact with study authors to identify additional studies) in the search and date last searched. Search 8 Present full electronic search strategy for at least one database, including any limits used, such that it could be repeated. Study selection 9 State the process for selecting studies (i.e., screening, eligibility, included in systematic review, and, if applicable, included in the meta-analysis). Data collection process 10 Describe method of data extraction from reports (e.g., piloted forms, independently, in duplicate) and any processes for obtaining and confirming data from investigators. Data items 11 List and define all variables for which data were sought (e.g., PICOS, funding sources) and any assumptions and simplifications made. Risk of bias in individual 12 Describe methods used for assessing risk of bias of individual studies (including specification of whether this was studies done at the study or outcome level), and how this information is to be used in any data synthesis. Summary measures 13 State the principal summary measures (e.g., risk ratio, difference in means). Synthesis of results 14 Describe the methods of handling data and combining results of studies, if done, including measures of c onsistency (e.g., I2) for each meta-analysis.

Page 1 of 2 PRISMA 2009 Checklist

Reported Section/topic # Checklist item on page # Risk of bias across studies 15 Specify any assessment of risk of bias that may affect the cumulative evidence (e.g., publication bias, selective reporting within studies). Additional analyses 16 Describe methods of additional analyses (e.g., sensitivity or subgroup analyses, meta-regression), if done, indicating which were pre-specified. RESULTS Study selection 17 Give numbers of studies screened, assessed for eligibility, and included in the review, with reasons for exclusions at each stage, ideally with a flow diagram. Study characteristics 18 For each study, present characteristics for which data were extracted (e.g., study size, PICOS, follow-up period) and provide the citations. Risk of bias within studies 19 Present data on risk of bias of each study and, if available, any outcome level assessment (see item 12). Results of individual studies 20 For all outcomes considered (benefits or harms), present, for each study: (a) simple summary data for each intervention group (b) effect estimates and confidence intervals, ideally with a forest plot. Synthesis of results 21 Present results of each meta-analysis done, including confidence intervals and measures of consistency. Risk of bias across studies 22 Present results of any assessment of risk of bias across studies (see Item 15). Additional analysis 23 Give results of additional analyses, if done (e.g., sensitivity or subgroup analyses, meta-regression [see Item 16]). DISCUSSION Summary of evidence 24 Summarize the main findings including the strength of evidence for each main outcome; consider their relevance to key groups (e.g., healthcare providers, users, and policy makers). Limitations 25 Discuss limitations at study and outcome level (e.g., risk of bias), and at review-level (e.g., incomplete retrieval of identified research, reporting bias). Conclusions 26 Provide a general interpretation of the results in the context of other evidence, and implications for future research. FUNDING Funding 27 Describe sources of funding for the systematic review and other support (e.g., supply of data); role of funders for the systematic review.

From: Moher D, Liberati A, Tetzlaff J, Altman DG, The PRISMA Group (2009). Preferred Reporting Items for Systematic Review s and Meta-Analyses: The PRISMA Statement. PLoS Med 6(7): e1000097. doi:10.1371/journal.pmed1000097 For more information, visit: www.prisma-statement.org. Page 2 of 2 The effects of toluene exposure on growth: a meta-analysis

Supplementary Table 1 – Search strings utilised

Inhalant abuse, adolescence, weight Toluene, adolescence, obesity Inhalant abuse, adolescence, height Toluene, adolescence, underweight Inhalant abuse, adolescence, BMI Toluene, adolescence, cachexia Inhalant abuse, adolescence, emaciation Toluene, adolescence, wasting Inhalant abuse, adolescence, malnutrition Toluene, adolescence, failure to thrive Inhalant abuse, adolescence, overweight Toluene, adolescence, length Inhalant abuse, adolescence, obesity Toluene, adolescence, spine length Inhalant abuse, adolescence, underweight Toluene, adolescence, stunting Inhalant abuse, adolescence, cachexia Toluene, adolescence, linear growth Inhalant abuse, adolescence, wasting Toluene, adolescence, waist circumference Inhalant abuse, adolescence, failure to thrive Toluene, adolescence, z score Inhalant abuse, adolescence, length Toluene, adolescence, height to weight ratio Inhalant abuse, adolescence, spine length Petrol glue sniffing, adolescence, weight Inhalant abuse, adolescence, stunting Petrol glue sniffing, adolescence, height Inhalant abuse, adolescence, linear growth Petrol glue sniffing, adolescence, BMI Inhalant abuse, adolescence, waist circumference Petrol glue sniffing, adolescence, emaciation Inhalant abuse, adolescence, z score Petrol glue sniffing, adolescence, malnutrition Inhalant abuse, adolescence, height to weight ratio Petrol glue sniffing, adolescence, overweight Volatile solvent abuse, adolescence, weight Petrol glue sniffing, adolescence, obesity Volatile solvent abuse, adolescence, height Petrol glue sniffing, adolescence, underweight Volatile solvent abuse, adolescence, BMI Petrol glue sniffing, adolescence, cachexia Volatile solvent abuse, adolescence, emaciation Petrol glue sniffing, adolescence, wasting Volatile solvent abuse, adolescence, malnutrition Petrol glue sniffing, adolescence, failure to thrive Volatile solvent abuse, adolescence, overweight Petrol glue sniffing, adolescence, length Volatile solvent abuse, adolescence, obesity Petrol glue sniffing, adolescence, spine length Volatile solvent abuse, adolescence, underweight Petrol glue sniffing, adolescence, stunting Volatile solvent abuse, adolescence, cachexia Petrol glue sniffing, adolescence, linear growth Volatile solvent abuse, adolescence, wasting Petrol glue sniffing, adolescence, waist circumference Volatile solvent abuse, adolescence, failure to thrive Petrol glue sniffing, adolescence, z score Volatile solvent abuse, adolescence, length Petrol glue sniffing, adolescence, height to weight ratio Volatile solvent abuse, adolescence, spine length Huffing Volatile solvent abuse, adolescence, stunting Volatile solvent abuse and adolescence Volatile solvent abuse, adolescence, linear growth Inhaled toluene and adolescence Volatile solvent abuse, adolescence, waist circumference Petrol sniffing and adolescence Volatile solvent abuse, adolescence, z score Toluene, food, appetite Volatile solvent abuse, adolescence, height to weight ratio Toluene and appetite Toluene, adolescence, weight Toluene and motor syndrome Toluene, adolescence, height Inhaled toluene and reproduction Toluene, adolescence, BMI Inhaled toluene and body weight Toluene, adolescence, emaciation Inhaled toluene and height Toluene, adolescence, malnutrition Inhaled toluene and body mass index Toluene, adolescence, overweight Inhaled toluene and bone

The effects of toluene exposure on growth: a meta-analysis

Supplementary Table 2 – summary results of risk of bias assessment

Unclear Item Type of bias Domain Review authors judgment No (n) (n) Yes (n) Total Was the allocation sequence adequately 1 Selection bias Sequence generation 4 13 14 31 generated and applied? Were the groups similar at baseline or 2 Selection bias Baseline characteristics where they adjusted for confounders in the 3 5 23 31 analysis?

3 Selection bias Allocation concealment Was the allocation adequately concealed? 4 27 0 31 Performance Were the animals randomly housed during 4 Random housing 7 23 1 31 bias the experiment? Were the caregivers and/or investigators Performance blinded from knowledge which 5 Blinding 5 26 0 31 bias intervention animals received during the experiment? Random outcome Were animals selected at random for 6 Detection bias 1 6 24 31 assessment outcome assessment? 7 Detection bias Blinding Was the outcome assessor blinded? 0 5 26 31 Were incomplete outcome data adequately 8 Attrition bias Incomplete outcome data 0 0 31 31 addressed? Selective outcome Are reports of the study free of selective 9 Reporting bias 31 0 0 31 reporting outcome reporting? Was the study apparently free of other 10 Other Other sources of bias problems that could result in a high risk of 5 0 26 31 bias?

Rose Crossin (737900)

Chapter 4 – The energy balance consequences of adolescent inhalant abuse and potential causal mechanisms Chapters 2 and 3 showed that adolescent inhalant abuse resulted in impairments to both weight and height, and that height impairments persisted into sustained abstinence. However, the effect of inhalants on food intake and other energy balance variables, and how they related to the observed growth changes, could not be assessed with the available data. Therefore, the first aim of this study was to characterise the effects of adolescent inhalant abuse in relation to components of the energy balance equation, and to determine the persistence of these effects into sustained abstinence. The rationale for this aim was that drugs of abuse have been shown to affect all variables within the energy balance equation (which is discussed in a review published and included as Appendix 1 to this thesis); therefore, characterisation should not just be limited to food intake. Furthermore, the long-term health effects of inhalant abuse cannot be determined unless it is known how changes persist or resolve in abstinence. The second aim of this study was to attempt to identify a mechanism underlying the observed growth effects, including specifically testing the hypotheses that reduced food intake (undernutrition) was the primary driver of growth impairments. Identification of a mechanism would provide the potential for future therapeutic approaches to be developed, to treat the growth impairments. The majority of this work has been accepted for publication in Neuroendocrinology in a manuscript titled: Adolescent inhalant abuse results in adrenal dysfunction and hypermetabolic phenotype with persistent growth impairments. The following formatting is consistent with the journal requirements and current status of the manuscript, which is not yet in-press. Additional data that were not included in this manuscript are provided as supplementary information to this chapter, after the accepted manuscript.

71 Rose Crossin

1 Adolescent inhalant abuse results in adrenal dysfunction and a hypermetabolic

2 phenotype with persistent growth impairments

3 Short title: Metabolic consequences of adolescent inhalant abuse

4 Rose Crossina, b, Zane B Andrewsc, Natalie A Simsd, Terence Panga, Michael Mathaie,

5 Jonathan H Gooif, Aneta Stefanidisc, Brian J Oldfieldc, Andrew J Lawrencea, g, Jhodie R

6 Duncana, h

7

8 aAddiction Neuroscience, Florey Institute of Neuroscience and Mental Health, Melbourne,

9 VIC, Australia, 3052

10 bEastern Health Clinical School, Monash University, Box Hill, VIC, Australia, 3128

11 cMonash Biomedicine Discovery Institute, Department of Physiology, Monash University,

12 Clayton, VIC, Australia, 3183

13 dSt Vincent’s Institute of Medical Research, and Department of Medicine at St. Vincent’s

14 Hospital, The University of Melbourne, Melbourne, VIC, Australia, 3065

15 eVictoria University, Melbourne, VIC, Australia, 3021

16 fThe University of Melbourne, Department of Medicine at St. Vincent’s Hospital, Fitzroy,

17 VIC, Australia, 3065

18 gFlorey Department of Neuroscience and Mental Health, The University of Melbourne,

19 Melbourne, VIC, Australia, 3010

20 hSchool of Medicine, University of Adelaide, Adelaide, SA, Australia, 5005

21

Page 1 of 48

Rose Crossin

1

2 Correspondence to:

3 Rose Crossin (ORCID ID https://orcid.org/0000-0003-1814-1330)

4 Florey Institute of Neuroscience and Mental Health

5 Parkville, Victoria 3052, Australia

6 Contact: +61 3 9035 6669 / [email protected]

7

8 Key words: toluene, volatile solvent abuse, hypo-cortisolism, negative energy balance,

9 abstinence, adrenal insufficiency

10

11 Disclosures and funding

12 The research was supported by the Australian National Health and Medical Research Council

13 (NHMRC) (940835), of which AJL is a Principal Research Fellow (1116930), ZBA is a

14 Career Development Fellow (1084344), and NAS was a Senior Research Fellow (1019803),

15 the Australian Dept. of Education and Training, from whom RC receives a scholarship

16 through the Research Training Program, and the Victorian Government’s Operational

17 Infrastructure Support Scheme. Funding bodies had no involvement in the design, analysis

18 and decision to publish. There are no conflicts of interest or financial disclosures in this work.

19

Page 2 of 48

Rose Crossin

1 Abstract

2 Background/aims: Abuse of toluene products (e.g. glue-sniffing) primarily occurs during

3 adolescence and has been associated with appetite suppression and weight impairments.

4 However, the metabolic phenotype arising from adolescent inhalant abuse has never been

5 fully characterised, and its persistence during abstinence and underlying mechanisms remain

6 unknown.

7 Methods: Adolescent male Wistar rats (PND 27) were exposed to inhaled toluene

8 (10,000ppm) (n=32) or air (n=48) for 1 hour/day, 3 days/week for 4 weeks, followed by 4

9 weeks abstinence. Twenty air-rats were pair-fed to the toluene group, to differentiate the

10 direct effects of toluene from under-nutrition. Food intake, weight and growth were

11 monitored. Metabolic hormones were measured after exposure and abstinence periods.

12 Energy expenditure was measured using indirect calorimetry. Adrenal function was assessed

13 using adrenal histology and hormone testing.

14 Results: Inhalant abuse suppressed appetite and increased energy expenditure. Reduced

15 weight gain and growth were observed in both toluene and pair-fed groups. Compared to the

16 pair-fed group, and despite normalisation of food intake, the suppression of weight and

17 growth for toluene-exposed rats persisted during abstinence. After exposure, toluene rats had

18 low fasting blood glucose and insulin compared to air and pair-fed groups. Consistent with

19 adrenal insufficiency, adrenal hypertrophy and increased basal adrenocorticotropic hormone

20 was observed in toluene-exposed rats, despite normal basal corticosterone levels.

21 Conclusions: Inhalant abuse results in negative energy balance, persistent growth impairment,

22 and endocrine changes suggestive of adrenal insufficiency. We conclude that adrenal

23 insufficiency contributes to the negative energy balance phenotype, potentially presenting a

24 significant additional health risk for inhalant users.

Page 3 of 48

Rose Crossin

1 Precis

2 Utilising an animal model replicating human abuse patterns, we found that adolescent

3 inhalant abuse induces growth impairments and a negative energy balance, attributable to

4 endocrine dysfunction.

Page 4 of 48

Rose Crossin

1 Introduction

2 Inhalant abuse involves inhalation of products containing toluene (e.g. glue, petrol, aerosols)

3 to induce intoxication; it often occurs intermittently over multiple years, resulting in chronic

4 exposure to toluene [1]. The concentration of toluene inhaled during a “session” typically

5 ranges from 3,000-15,000 parts per million (ppm) [2]. Inhalant abuse is predominantly

6 associated with early adolescence [3, 4], thus the peak period of exposure to toluene overlaps

7 with the adolescent growth spurt; characterised by a rapid increase in body weight and height,

8 around 12-15 years of age in males [5]. In humans, inhalant abuse has been associated with

9 emaciation [6], weight impairment [7], and reduced height trajectory persisting into sustained

10 abstinence [7, 8]. However, the mechanisms underlying these growth impairments remain

11 unknown.

12 Inhalant abuse reduces food intake in humans [9] and animal models [10, 11]. However,

13 evidence that under-nutrition is the sole mediator of inhalant-induced growth changes is

14 limited. Dick et al (2016) showed exposure to toluene during adolescence alters the

15 relationship between food intake and body weight, and caused metabolic dysfunction

16 including non-diabetic fasting hypoglycaemia and decreased adiposity, suggestive of a

17 broader energy balance effect [10]. Energy balance is a function of energy intake (moderated

18 by absorption) less energy expended, with a negative energy balance when expenditure

19 exceeds intake. Despite the knowledge of lack of weight gain from inhalant abuse, the energy

20 balance effects remain unknown. Thus, our aim was to characterise the growth and metabolic

21 consequences of adolescent inhalant abuse during exposure and abstinence, using the energy

22 balance equation and a pair-feeding model to determine the role of under-nutrition.

23 Another factor implicated in growth changes following inhalant abuse is non-diabetic fasting

24 hypoglycaemia [10]. This rare condition is generally related to conditions such as renal

Page 5 of 48

Rose Crossin

1 insufficiency, liver disease, or adrenal insufficiency [12, 13]. Preliminary evidence supports a

2 hypothesis that adolescent inhalant abuse results in adrenal insufficiency, since adrenal

3 hypertrophy has been reported in animal models [10, 14, 15]. Adrenal insufficiency is a

4 failure to produce adequate levels of cortisol (corticosterone in rodents), during basal or high-

5 stress conditions [16]. Though cortisol is produced by the adrenal glands, the signalling

6 pathway derives from the hypothalamic-pituitary-adrenal (HPA) axis, thus insufficient

7 cortisol can be caused by a primary insufficiency in the adrenal glands or a secondary

8 insufficiency in the hypothalamus or pituitary. Symptoms of adrenal insufficiency include

9 weight loss, growth impairments and reduced growth trajectory, non-diabetic fasting

10 hypoglycaemia, nausea and gastric pain, persistent low-grade fever, and fatigue [16]. In

11 isolation, all these symptoms have been associated with inhalant abuse [6-10, 17-19]

12 however, whether adrenal insufficiency follows inhalant abuse has never been formally

13 tested. Thus, our second aim was to test whether adrenal insufficiency may contribute to the

14 metabolic and growth consequences of inhalant exposure, using a process to diagnose adrenal

15 insufficiency and localise it within the HPA-axis (Figure 1).

16

Page 6 of 48

Rose Crossin

1 Methods and Materials

2 Animals

3 Adolescent male Wistar rats post-natal day (PND) 24 (n=80) (Animal Resource Centre,

4 Perth, Western Australia) were maintained on a 12-hour light/dark cycle (7am/7pm) and

5 singly housed to monitor ad libitum food (standard chow) and water intake for Air-control

6 and chronic intermittent toluene (CIT) groups, with a pair-fed (to CIT animals) control group

7 used to differentiate the direct effects of toluene from the secondary effects of under-

8 nutrition. The food intake calculation for the pair-fed group is provided (Supplementary File

9 1). Rats were acclimated for a minimum of 2 days prior to experimentation. Experiments

10 were performed in accordance with the Prevention of Cruelty to Animals Act 1986 under the

11 guidelines of the Australian National Health and Medical Research Council Code of Practice

12 for the Care and Use of Animals for Experimental Purposes in Australia and in compliance

13 with institutional, national and international guidelines, approved by the Florey animal ethics

14 committee (March 2013, 13-021).

15 Chronic intermittent toluene (CIT) procedure

16 Rats were exposed to CIT as described [20]. Briefly, exposure was undertaken in toluene-

17 resistant chambers (17.6 cm wide × 16.5 cm high × 16.4 cm deep; Alternative Plastics Pty

18 Ltd, North Melbourne, Australia; fittings by Swagelok, Broadmeadows, Australia) with

19 toluene concentration maintained at 10,000±100 ppm (verified by calibrated inline gas

20 chromatography; Shimadzu Corporation, Kyoto, Japan). Equivalent chambers exposed to

21 room air (0 ppm toluene) were used for Air-control and pair-fed controls.

22 Rats were randomly assigned to Air (control n=28, or pair-fed control n=20) or 10,000 ppm

23 toluene (CIT) (n=32) for 1 hr/day, 3 days/week, for 4 weeks. A sub-group of rats were killed

Page 7 of 48

Rose Crossin

1 for tissue collection after the 4 week exposure period (Air-control n=10, Pair-fed n=10, CIT

2 n=11), while the remainder (Air-control n=18, Pair-fed n=10, CIT n=21) experienced 4

3 weeks abstinence. Exposure was conducted at the same time each day at room temperature

4 (21°C). Not all animals underwent all tests; numbers per group are described for each test.

5 Tissue collection

6 Following experimentation, animals were euthanized (Lethabarb overdose 1mL/kg i.p.

7 followed by decapitation). The brain, liver, heart, lung, right kidney, right adrenal gland, right

8 testis, and the epididymal fat pad (eWAT) were dissected and weighed. The left adrenal gland

9 was dissected and preserved in 10% neutral buffered formalin. Hindlegs were dissected at the

10 hip-joint, excess tissue removed, and fixed in 4% paraformaldehyde for 24 hours, then

11 transferred to 70% ethanol.

12 Food/water intake and growth measures

13 Body weight, food and water intake were measured 3 days/week. Rump width was measured

14 weekly using callipers, at the widest point, when the rat was in a non-extended stance with

15 hind-legs under the rump. Spine length was measured weekly from the base of skull to top of

16 the tail. Measures were taken throughout exposure and abstinence (Air n=10, Pair-fed n=10,

17 CIT n=12).

18 Bomb calorimetry

19 Bomb calorimetry was utilised to measure faecal energy content at the end of exposure (Air-

20 control n=20, Pair-fed n=20, CIT n=23) and abstinence (Air-control n=10, Pair-fed n=10,

21 CIT n=12). Faeces were collected over a 24-hr period and stored at -80°C. Faeces were

22 weighed, dried (60°C), ground to a powder and transferred to gelatin caps (size 0; approx. 0.1

23 g faeces/cap). Bomb calorimetry was conducted in an Cal2k calorimeter (DDS calorimeters,

Page 8 of 48

Rose Crossin

1 South Africa) with empty gelatin caps used as blanks, duplicates for each sample, and

2 calibration using 0.5g benzoic acid tablets (DDS calorimeters, South Africa).

3 3-bottle choice

4 A preference test between water, sucrose (2% w/v), and saccharin (0.1% w/v) [21] was

5 performed after exposure (Air-control n=20, Pair-fed n=20, CIT n=23) and abstinence (Air-

6 control n=10, Pair-fed n=10, CIT n=12). Throughout the exposure/abstinence protocol

7 animals were provided with 3 water bottles, to prevent novel object exploration during the 3-

8 bottle choice. Bottle position was randomised; bottles were weighed once/day, to provide for

9 daily consumption, over a 3-day period.

10 Metabolic cages

11 Indirect calorimetry was utilised at the end of exposure (Air-control n=8, CIT n=8) to

12 measure oxygen consumption and carbon dioxide production. Additionally, energy

13 expenditure and respiratory exchange ratio were determined, as described [22]. Animals were

14 housed within enclosed calorimetry chambers (LabMaster; TSE-systems, Bad Homburg,

15 Germany) for 48 hours (baseline), followed by an 18 hour fast (water was available ad

16 libitum), then 6 hour re-feeding period.

17 Circulating metabolic hormones and liver glycogen

18 Insulin, leptin and blood glucose were measured sated and following a 12 hour dark cycle fast

19 at the end of exposure (Air-control n=20, Pair-fed n=20, CIT n=23) and abstinence (Air-

20 control n=10, Pair-fed n=10, CIT n=12). Blood glucose was measured using a tail snip

21 (Accu-Chek® Active blood glucose meter) and 300 µL blood was collected in BD800 blood

22 metabolic preservation tubes (BD, North Ryde, NSW, Australia), spun (8,400 g, 4°C, 15

23 mins) and plasma stored at -80°C. Plasma was tested for insulin (Crystal Chem 90080) and

Page 9 of 48

Rose Crossin

1 leptin (EMD Millipore EZRL-83K) by ELISA kits. Results were read on a BioTek EPOCH2

2 microplate reader.

3 Glycogen was tested using 10 mg of liver collected at the end of exposure (Air-control n=10,

4 Pair-fed n=10, CIT n=11) and abstinence (Air-control n=10, Pair-fed n=10, CIT n=12).

5 Samples were homogenised and the supernatant prepared as per manufacturer instructions,

6 with analysis by ELISA kit (abcam ab65620) and read as above.

7 Skeletal microcomputed tomography (µCT) and mechanical testing

8 Tibiae were analysed by µCT at the end of exposure (Air-control n=10, Pair-fed n=10, CIT

9 n=11) and abstinence (Air-control n=10, Pair-fed n=10, CIT n=12) using the SkyScan 1076

10 (Bruker-microCT, Kontich, Belgium) [23, 24]. Images were acquired using the following

11 settings: 9 μm voxel resolution, 0.5 mm aluminium filter, 69 kV voltage, 139 μA current,

12 1950 ms exposure time, rotation 0.5°, frame averaging=1. The images were reconstructed and

13 analysed using NRecon (1.6.10.2), DataViewer (1.4.4), and CT Analyser (1.14.4.1) [23]. The

14 trabecular region of interest (ROI) was a 4.5 mm region starting 3 mm below the proximal

15 growth plate. Cortical ROI was a 4.5 mm region starting 7 mm below the growth plate, such

16 that the middle of the ROI corresponded with the point of loading used for three-point

17 bending tests. Bone structure analysis was completed with global thresholding in CTAn

18 (1.14.4.1). Thresholds were set at a lower threshold of 0.632 mg/cm3 CaHA for trabecular

19 bone and 0.825 mg/cm3 CaHA for cortical bone, based on multilevel Otsu thresholding of the

20 entire dataset. Tissue mineral density was quantified as described [25]. To determine bone

21 mechanical properties at the end of the abstinence period (Air n=10, Pair-fed n=10, CIT

22 n=12), 3-point bending was conducted as described [24] (Supplementary File 2). Structural

23 properties were calculated [23, 26] in combination with morphological data from μCT [27].

24 Adrenal histology

Page 10 of 48

Rose Crossin

1 To identify adrenal hypertrophy, fixed adrenal glands collected at the end of exposure (Air-

2 control n=10, CIT n=11) and abstinence (Air-control n=10, CIT n=12) were paraffin-

3 embedded, sectioned (5µm) and stained with hematoxylin and eosin (H&E). For each animal,

4 the width of each zone of the adrenal gland was measured 4 times across 3 sections, yielding

5 12 measurements. Within each zone, individual cells were measured: 10/zone, across 3

6 sections, yielding 30 measurements. Measurements were conducted blinded, using ImageJ

7 (Version 1.49b National Institutes of Health, USA).

8 Basal corticosterone

9 Basal corticosterone was measured sated and following a 12 hour dark cycle fast at the end of

10 exposure (Air-control n=20, CIT n=23) and abstinence (Air-control n=10, CIT n=12). Sated

11 blood samples were collected between 9 am and 11 am, food was then removed at 9 pm to

12 provide a 12 hour fast throughout the dark cycle, and fasted blood samples were collected

13 between 9 am and 11 am the following day. Approximately 300 µL blood was collected in

14 BD800 blood metabolic preservation tubes (BD, North Ryde, NSW, Australia), spun (8,400

15 g, 4°C, 15 mins) and plasma stored at -80°C. Plasma was tested for corticosterone by ELISA

16 kit (Abcam ab108821) and results read on a BioTek EPOCH2 microplate reader.

17 Insulin tolerance test (ITT)

18 An ITT was performed at the end of the exposure period (Air-control n=8, CIT n=9). Insulin

19 (0.5U/kg i.p.) was administered and blood glucose was measured at baseline, and 15, 30, 45,

20 60 and 90 minutes post-injection (Accu-Chek® Active blood glucose meter). At baseline and

21 30, 60 and 90 mins post-injection 300 µL blood was collected and plasma tested for

22 corticosterone, as above.

23 Stress response test

Page 11 of 48

Rose Crossin

1 A 30-minute restraint stress test was performed at the end of the exposure period (Air-control

2 n=8, CIT n=9). Blood glucose was measured at baseline, and 0, 15, 30, 45, 60 and 90 minutes

3 post-restraint (Accu-Chek® Active blood glucose meter). Approximately 300µL blood was

4 obtained via tail snips (baseline and 0, 30, 60 and 90 minutes post-restraint), as above.

5 Plasma was tested for corticosterone, as above, and adrenocorticotropic hormone (ACTH)

6 (Phoenix Pharmaceuticals EK-001-21).

7 Statistical analysis

8 Statistical analysis was performed using SPSS (IBM Version 22). Repeated measures were

9 analysed as a two-way ANOVA over time, or single measures as a one-way ANOVA (all 3

10 groups), with post-hoc corrections for multiple group comparisons. Comparisons between

11 single measures for two groups were analysed with an independent samples t-test. Details of

12 statistical tests for each variable are in Supplementary File 3. Results are reported as

13 mean±standard error of the mean (SEM) and considered significant at p<0.05.

14

15

16

17

Page 12 of 48

Rose Crossin

1 Results

2 Inhalant abuse causes persistent disruptions to energy balance

3 Food intake

4 To investigate the energy intake side of the energy balance equation, food consumption and

5 absorption were assessed. Daily food intake in chronic intermittent toluene (CIT) and Pair-

6 fed groups was significantly lower than Air-controls from the third exposure day onwards,

7 and was reduced by 26% at the end of exposure (F=12.064 p=0.001, Figure 2A), noting that

8 these results were not significant when corrected by body weight (Supplementary Figure 1A)

9 or metabolic mass at power of 0.75 (data not shown). In the CIT group, food intake

10 normalised to Air-control by day 15 of abstinence (F=3.944 p=0.031, Figure 2A). Cumulative

11 food intake in CIT and Pair-fed groups was significantly lower than Air-control at the end of

12 exposure (F=7.410 p=0.003) and abstinence (F=9.403 p=0.001). See Supplementary File 4

13 for additional data of food intake per gram body weight, water intake, and conversion of food

14 intake to body weight.

15 Food intake differences were not moderated by differences in energy absorption, with faecal

16 bomb calorimetry revealing no differences between groups at the end of exposure or

17 abstinence. There were no group differences for preference between saccharin or sucrose at

18 the end of both exposure and abstinence, though all groups consumed more saccharin and

19 sucrose than water at the end of exposure (liquid type F=43.27 p<0.0001) and abstinence

20 (liquid type F=21.52 p<0.0001).

21 Body weight and linear growth

22 To characterise effects of toluene on growth, body weight, spine length, and rump width were

23 measured. Body weight in the CIT group was significantly lower than Air-control and Pair-

Page 13 of 48

Rose Crossin

1 fed groups from exposure day 2; by the end of exposure both the CIT and Pair-fed groups

2 were significantly lower than Air-control. After 2 weeks of abstinence, the Pair-fed group

3 normalised to Air-control, whereas the CIT group remained significantly lower than both the

4 Air-control and Pair-fed at the end of abstinence (F=9.922 p=0.001, Figure 2B). Spine length

5 (F=9.879 p=0.001, Figure 2C) and rump width (F=3.817 p=0.034, Figure 2D) were

6 significantly lower than Air-control at the end of exposure for CIT and Pair-fed animals, but

7 only the CIT remained significantly lower after abstinence for both spine length (F=23.065

8 p<0.0001, Figure 2C) and rump width (F=3.932 p=0.031, Figure 2D).

9 Indirect calorimetry

10 To establish whether there were changes in energy expenditure side in response to toluene

11 treatment, indirect calorimetry was completed. At the end of exposure, CIT animals showed

12 elevated baseline metabolic rate compared to Air-control (F=8.602 p=0.0109, Figure 3A).

13 Similarly, baseline energy expenditure was higher in CIT animals (F=9.108 p=0.0092, Figure

14 3B). For both baseline metabolic rate (F=4.67 p=0.0210) and baseline energy expenditure

15 (F=4.976 p=0.0170) the increase in the CIT group was greater compared to Air-control in the

16 dark cycle. There were no significant group differences in baseline respiratory exchange ratio

17 (RER) or RER in the fasted state, but CIT animals had higher RER during a re-feeding period

18 following a fast (F=10.08 p=0.0068, Figure 3C), indicative of a preferential increase in the

19 oxidation of carbohydrates. There were no group differences in food intake in either

20 paradigm (Figures 3D and 3E), similarly no group differences in baseline water intake,

21 though the CIT animals did consume less water during a re-feeding period following a fast

22 (t=2.261 p=0.0402, Figure 3F). Additional metabolic variables are provided in

23 Supplementary File 5.

24 Metabolic hormones

Page 14 of 48

Rose Crossin

1 To test for metabolic perturbations, hormone levels were tested in sated and fasted

2 conditions. At the end of exposure the CIT group showed reductions in fasted blood glucose

3 levels (F=7.686 p<0.0001, Figure 4A) and fasted insulin (F=4.158 p=0.0204, Figure 4B),

4 with no difference in liver glycogen (Figure 4C). Both the Pair-fed and CIT groups had

5 significantly reduced leptin levels compared to Air-control animals in both sated and fasted

6 states (F=8.339 p=0.0004, Figure 4D). There were no significant group differences in blood

7 glucose, insulin, leptin, or liver glycogen at the end of abstinence in sated or fasted states

8 (data not shown).

9 Skeletal microCT and mechanical testing

10 As there was a decrease in linear growth rate in the CIT group, skeletal structure was

11 assessed. At the end of exposure, the Pair-fed group compared to both the Air-control and

12 CIT groups showed low trabecular bone volume (F=4.783 p=0.0167, Figure 5A) and

13 trabecular number (F=5.342 p=0.0111, Figure 5C), but not thickness (Figure 5B), and greater

14 trabecular separation (F=8.167 p=0.0017, Figure 5D). Trabecular structure did not differ

15 between CIT and Air-control, nor between any groups at the end of abstinence (Figure 5A-

16 D). In contrast, at the end of abstinence the CIT group had lower cortical thickness than Air-

17 control and Pair-fed (F=11.89 p=0.0002, Figure 5E). No group differences were observed for

18 tissue mineral density at either time point (Figure 5F). Despite lower cortical thickness in the

19 CIT group at the end of abstinence, there were no group differences in bone mechanical

20 properties (including Young’s Modulus, Yield Stress, Ultimate Stress and Failure Stress).

21 Additional skeletal µCT and bone mechanical property variables are provided in

22 Supplementary File 6.

23 Adolescent inhalant abuse promotes adrenal insufficiency

Page 15 of 48

Rose Crossin

1 For all further experiments pair-fed groups were excluded, as results indicated under-nutrition

2 was not a primary driver of the observed growth and metabolic phenotype.

3 Adrenal hypertrophy

4 To begin the diagnostic process for adrenal insufficiency, adrenal histology was conducted.

5 Post-mortem organ weights are reported in Table 1. Adrenal glands were heavier in the CIT

6 group, when corrected for body weight at end of exposure (F=7.300 p=0.003). At end of

7 abstinence, livers were heavier in the CIT group, when corrected for body weight (F=11.865

8 p<0.0001). There were no differences in the other organ weights collected.

9 Adrenal histology, basal plasma corticosterone, and ACTH levels

10 The CIT group had increased zona fasiculata zone width (exposure t=2.684 p=0.0152,

11 abstinence t=5.293 p<0.0001, Figure 6A) and cell size width (exposure t=6.334 p<0.0001,

12 abstinence t=5.341 p<0.0001, Figure 6B) at the end of both exposure and abstinence (i.e.

13 Figure 1 box 1). Additional measures are provided in Supplementary File 7. To differentiate

14 between stress and adrenal insufficiency in the context of the observed adrenal hypertrophy,

15 basal corticosterone and ACTH levels were assessed. Despite a difference in adrenal weight

16 and histology there were no significant group differences in basal corticosterone levels at the

17 end of exposure, in either the sated or fasted state (Figure 6C), however basal corticosterone

18 levels were decreased in the CIT animals compared to Air-control, at the end of abstinence,

19 in the sated state (t=2.172 p=0.042, Figure 6D) (i.e. Figure 1 box 3). Increased levels of basal

20 ACTH were observed in the CIT group compared to Air-control, at the end of exposure

21 (t=2.806 p=0.0133, Figure 6E) (i.e. Figure 1 box 4 – primary adrenal insufficiency).

22 Adrenal function

Page 16 of 48

Rose Crossin

1 To test for secondary or tertiary adrenal insufficiency, and to assess adrenal function, an

2 insulin tolerance test (ITT) and stress response test were conducted. At the end of exposure

3 an ITT showed an increased response in both blood glucose (F=9.766 p=0.0075, Figure 7A)

4 and corticosterone (F=30.09 p<0.0001, Figure 7B) in CIT animals between 30-60 minutes

5 post-insulin administration, compared to Air-control (i.e. Figure 1 box 5). Following restraint

6 stress the CIT group showed elevated baseline ACTH levels (t=2.806 p=0.0133) but did not

7 show a strong rise in ACTH after the stressor, compared to Air-control (Figure 7C). There

8 was no significant group difference in corticosterone levels through the stress response test

9 until 90 minutes post-stress, where CIT animals had significantly elevated corticosterone

10 compared to Air-control (t=2.968 p=0.019, Figure 7D). The CIT animals had significantly

11 lower blood glucose levels at Time 0 within the stress response test (t=3.166 p=0.007, Figure

12 7E), with no differences at any other time point. The differentials for change in blood glucose

13 for each unit change in corticosterone within the stress response test are shown in Figure 8.

14 The blood glucose response to changing corticosterone levels, within the context of a stress

15 response test, was blunted in the CIT group compared to Air-control, with twice the amount

16 of change to corticosterone needed in the CIT group, to achieve the same change in blood

17 glucose.

18

Page 17 of 48

Rose Crossin

1 Discussion

2 Our study is the first, to our knowledge, to uncover a range of metabolic and endocrine

3 disturbances linked to impaired growth as a result of adolescent inhalant abuse, which is

4 sustained throughout abstinence. These include reduced food intake during exposure,

5 increased energy expenditure after exposure, and non-diabetic fasting hypoglycaemia. Pair-

6 feeding revealed both the growth impairment and metabolic disruption following adolescent

7 inhalant abuse are independent of decreased food intake. We showed for the first time, to our

8 knowledge, that adolescent inhalant abuse results in adrenal dysfunction, consistent with

9 adrenal insufficiency, which is non-recoverable by abstinence. Our data are diagnostically

10 consistent with primary adrenal insufficiency; presenting a potential significant chronic

11 health risk for individuals with a history of inhalant abuse, as adrenal insufficiency can be

12 life-threatening when undiagnosed and untreated [16].

13 Previous rat studies established that adolescent inhalant abuse decreases food intake and body

14 weight gain [10, 11], but while it has been suggested this involves dysregulation of appetite

15 stimulating hormones such as neuropeptide Y [11], the underlying cause remains unclear. We

16 have determined that these changes are not driven by under-nutrition since food intake

17 normalised during abstinence, with no over-consumption observed, which would be expected

18 following a period of under-nutrition [28, 29]. Instead, we found that there was a concurrent

19 increase in energy expenditure, including basal metabolic rate. Thus, there is a dual effect on

20 the energy balance equation. Moreover, substrate utilisation differed following inhalant

21 abuse, with a rapid return to carbohydrate utilisation following fasting. This represents an

22 energy conserving mechanism due to limited adipose stores, forcing a return to carbohydrate

23 utilisation at the expense of maintaining blood glucose levels, a process controlled by hunger-

24 sensing agouti-related peptide (AgRP) neurons in the hypothalamus [30]. Future studies

Page 18 of 48

Rose Crossin

1 investigating tissue-specific changes in energy expenditure and metabolism following chronic

2 toluene exposure would be valuable.

3 Through pair-feeding we found under-nutrition resulted in decreased body weight and growth

4 during the exposure period and decreased leptin levels [10]. However, CIT animals showed

5 non-diabetic fasting hypoglycaemia, which was not observed in the pair-fed group.

6 Additionally, despite food intake recovering in abstinence, body weight continued to be

7 suppressed [10] whereas body weight and growth in the pair-fed group rapidly normalised to

8 the Air-controls. Adolescent inhalant abuse has been previously associated with decreased

9 bone mineral density [31], however, we did not replicate this finding. We found the pair-fed

10 group had less trabecular bone mass following the exposure period which resolved, consistent

11 with under-nutrition [32], suggesting a broader metabolic disruption caused by inhalant

12 abuse. Further exploration of the effects of adolescent inhalant abuse on hormonal growth

13 factors, including growth hormone (GH) and insulin-like growth factor 1 (IGF-1), would be

14 beneficial to elucidate potential causes of the observed persistent growth impairment.

15 Having identified a persistent negative energy balance phenotype, independent of under-

16 nutrition, further causal hypotheses were considered. Non-diabetic fasting hypoglycaemia is

17 generally associated with a disease state, including renal and adrenal insufficiency [12, 13].

18 Our bone data also suggested adrenal involvement, because the effect of under-nutrition on

19 bone is an indirect endocrine effect, including increased cortisol [33-35]. Finally, we

20 observed adrenal hypertrophy in the inhalant group following exposure, a common outcome

21 of adrenal insufficiency [36]. Utilising a diagnostic flowchart (Figure 1) we observed that

22 following the exposure period, inhalant exposure resulted in adrenal hypertrophy, particularly

23 in the zona fasiculata, with unchanged basal corticosterone levels but increased basal ACTH

24 levels. This is diagnostically consistent with primary adrenal insufficiency (i.e. Figure 1 box

25 4) [16]. Indeed, two previous inhalant abuse studies reported similar findings, but concluded

Page 19 of 48

Rose Crossin

1 inhalant abuse resulted in a high-stress phenotype without consideration of adrenal

2 insufficiency [14, 15].

3 We also conducted an ITT to test for secondary/tertiary adrenal insufficiency. As a

4 corticosterone peak was observed in response to an ITT, our results were inconsistent with an

5 insufficiency occurring upstream (i.e. findings inconsistent with Figure 1 box 5). However,

6 CIT animals were more insulin-sensitive than Air-controls within the ITT, limiting

7 interpretation. Further studies into hypothalamic and pituitary function within this model

8 would be beneficial. We also tested the functionality of the HPA-axis via a stress-response

9 test at the end of exposure and found a blunted stress and glucose response. This is suggestive

10 of glucocorticoid resistance [37], potentially occurring simultaneously with adrenal

11 insufficiency. At the end of abstinence, adrenal hypertrophy remained and basal

12 corticosterone levels were decreased compared to Air-controls in the fasted state, again

13 suggestive of adrenal insufficiency, though it would be beneficial for future work to consider

14 stress responsivity and the potential for glucocorticoid resistance at this time point to

15 understand the persistence of the observed effects.

16 Adrenal insufficiency has never before been identified as a risk factor for adolescent inhalant

17 abuse, and requires clinical confirmation. We found a single clinical case report linking

18 inhalant abuse to an effect on the adrenal gland [38], but this has not been causally

19 established in humans. If confirmed, this would represent a significant health risk to

20 individuals with a history of inhalant abuse, particularly if adrenal insufficiency persists

21 following abstinence as our results suggest. The symptoms of adrenal insufficiency include

22 fatigue, weight less, nausea, and irritability, all of which can impair quality of life, however,

23 adrenal insufficiency can result in a life-threatening adrenal crisis, particularly during surgery

24 or trauma, and this presents a major health risk particularly for those with undiagnosed or

25 untreated adrenal insufficiency [16]. Furthermore, our finding of adrenal insufficiency

Page 20 of 48

Rose Crossin

1 provides a potential mechanistic link between inhalant abuse and subsequent suicidal

2 behaviour [39], which has been associated with decreased HPA axis responsivity [40, 41].

3 Conclusion

4 Since the 1960’s it has been noted that inhalant users present as underweight [6]. We found

5 inhalant abuse during adolescence decreased food intake with a concomitant increase in

6 energy expenditure, ultimately resulting in persistently impaired growth. Adolescent inhalant

7 abuse resulted in adrenal dysfunction suggestive of adrenal insufficiency. This is a chronic

8 disorder requiring life-long treatment in humans, with potentially life-threatening

9 complications [16]; a health risk requiring further research in individuals with a history of

10 inhalant abuse.

11

Page 21 of 48

Rose Crossin

1 Acknowledgements

2 Authors gratefully acknowledge the contribution of Narelle McGregor (St Vincent’s Institute)

3 for assistance with the µCT, Erika Greaves (Monash University) for assistance with the

4 indirect calorimetry experiments, Dr. Enda Crossin (Swinburne University) for assistance

5 with blood glucose differential calculations, and the Florey core histology service (Florey

6 Institute) for the preparation of adrenal slides.

7

Page 22 of 48

Rose Crossin

1 References

2 1. Lubman, D., M. Yücel, and A. Lawrence, Inhalant abuse among adolescents: neurobiological 3 considerations. British journal of pharmacology, 2008. 154(2): p. 316-326. 4 2. Bowen, S.E., et al., The last decade of solvent research in animal models of abuse: 5 mechanistic and behavioral studies. Neurotoxicology and teratology, 2006. 28(6): p. 636- 6 647. 7 3. Australian Institute of Health and Welfare, 2010 National Drug Strategy Household Survey 8 Report. 2011, Australian Institute of Health and Welfare: Canberra. 9 4. Cancer Council Victoria, Australian secondary school students' use of tobacco, alcohol, and 10 over-the-counter and illicit substances in 2011. 2012, Cancer Council of Victoria for 11 Australian Government Department for Health and Ageing: Melbourne. 12 5. Tanner, J. and R. Whitehouse, Clinical longitudinal standards for height, weight, height 13 velocity, weight velocity, and stages of puberty. Archives of disease in childhood, 1976. 14 51(3): p. 170-179. 15 6. Glaser, H.H. and O.N. Massengale, Glue-sniffing in children: Deliberate inhalation of 16 vaporized plastic cements. JAMA, 1962. 181(4): p. 300-303. 17 7. Crossin, R., et al., Adolescent inhalant abuse leads to other drug use and impaired growth; 18 implications for diagnosis. Australian and New Zealand journal of public health, 2016. 19 8. Crossin, R., et al., The persistence of growth impairments associated with adolescent inhalant 20 abuse following sustained abstinence. Addiction Research & Theory, 2017: p. 1-4. 21 9. Pisetsky, E.M., et al., Disordered eating and substance use in high‐school students: Results 22 from the Youth Risk Behavior Surveillance System. Int. J. Eat. Disord., 2008. 41(5): p. 464-470. 23 10. Dick, A., et al., Chronic intermittent toluene inhalation in adolescent rats results in metabolic 24 dysfunction with altered glucose homeostasis. British journal of pharmacology, 2015. 25 172(21): p. 5174-5187. 26 11. Morón, L., et al., Toluene alters appetite, NPY, and galanin immunostaining in the rat 27 hypothalamus. Neurotoxicology and teratology, 2004. 26(2): p. 195-200. 28 12. Virally, M. and P. Guillausseau, Hypoglycemia in adults. 1999. 29 13. Fajans, S.S. and J.C. Floyd Jr, Fasting hypoglycemia in adults. N. Engl. J. Med., 1976. 294(14): 30 p. 766-772. 31 14. Ishigami, A., et al., Immunohistochemical study of rat spermatogenesis after toluene- 32 inhalation. Legal Medicine, 2005. 7(1): p. 42-46. 33 15. Yang, M., et al., Toluene Induces Depression-Like Behaviors in Adult Mice. Toxicological 34 research, 2010. 26(4): p. 315. 35 16. Arlt, W. and B. Allolio, Adrenal insufficiency. The Lancet, 2003. 361(9372): p. 1881-1893. 36 17. Baydala, L., Inhalant abuse. Paediatr. Child Health, 2010. 15(7): p. 443-448. 37 18. Connors, N.J. Inhalants Clinical Presentation. 2017 [cited 2017 17 October 2017]; Available 38 from: https://emedicine.medscape.com/article/1174630-clinical. 39 19. Anderson, C.E. and G.A. Loomis, Recognition and prevention of inhalant abuse. Am. Fam. 40 Physician, 2003. 68(5): p. 869-874. 41 20. Duncan, J.R., et al., Adolescent toluene inhalation in rats affects white matter maturation 42 with the potential for recovery following abstinence. PloS one, 2012. 7(9): p. e44790. 43 21. Ueji, K., et al., Saccharin Taste Conditions Flavor Preference in Weanling Rats. Chem. Senses, 44 2015. 41(2): p. 135-141. 45 22. Stefanidis, A., et al., Prevention of the adverse effects of olanzapine on lipid metabolism with 46 the antiepileptic zonisamide. Neuropharmacology, 2017. 123: p. 55-66. 47 23. Johnson, R.W., et al., The primary function of gp130 signaling in osteoblasts is to maintain 48 bone formation and strength, rather than promote osteoclast formation. J. Bone Miner. Res., 49 2014. 29(6): p. 1492-1505.

Page 23 of 48

Rose Crossin

1 24. Williamson, L., et al., High dose dietary vitamin D 3 increases bone mass and strength in 2 mice. Bone reports, 2017. 6: p. 44-50. 3 25. Ansari, N., et al., Autocrine and Paracrine Regulation of the Murine Skeleton by Osteocyte‐ 4 Derived Parathyroid Hormone‐Related Protein. J. Bone Miner. Res., 2017. 5 26. Jepsen, K.J., et al., Establishing biomechanical mechanisms in mouse models: practical 6 guidelines for systematically evaluating phenotypic changes in the diaphyses of long bones. J. 7 Bone Miner. Res., 2015. 30(6): p. 951-966. 8 27. Turner, C.H. and D.B. Burr, Basic biomechanical measurements of bone: a tutorial. Bone, 9 1993. 14(4): p. 595-608. 10 28. Dulloo, A.G., J. Jacquet, and L. Girardier, Poststarvation hyperphagia and body fat 11 overshooting in humans: a role for feedback signals from lean and fat tissues. The American 12 journal of clinical nutrition, 1997. 65(3): p. 717-723. 13 29. Dulloo, A.G., Human pattern of food intake and fuel-partitioning during weight recovery 14 after starvation: a theory of autoregulation of body composition. Proc. Nutr. Soc., 1997. 56: 15 p. 25-40. 16 30. Reichenbach A, S.R., Mequinion M, Denis RRG, Goularte JF, Clarke RE, Lockie SH, Lemus MB, 17 Kowalski GM, Bruce CR, Schittenhelm R, Huang C, Mynatt R, Oldfield BJ, Watt MJ, Luquet S, 18 Andrews ZB AgRP neurons require carnitine acetyltransferase (Crat) to regulate metabolic 19 flexibility and peripheral nutrient partitioning. . Cell Reports, 2018. Accepted in press. 20 31. Dündaröz, M.R., S. Sarici, and T. Turkbay, Evaluation of bone mineral density in chronic glue 21 sniffers. Turk. J. Pediatr, 2002. 44: p. 326-329. 22 32. Compston, J., et al., Effect of diet-induced weight loss on total body bone mass. Clin. Sci., 23 1992. 82(4): p. 429-432. 24 33. Newman, M.M. and K.A. Halmi, Relationship of bone density to estradiol and cortisol in 25 anorexia nervosa and bulimia. Res., 1989. 29(1): p. 105-112. 26 34. Munoz, M. and J. Argente, Anorexia nervosa in female adolescents: endocrine and bone 27 mineral density disturbances. European Journal of Endocrinology, 2002. 147(3): p. 275-286. 28 35. Miller, K.K., Endocrine effects of anorexia nervosa. Endocrinol. Metab. Clin. North Am., 2013. 29 42(3): p. 515. 30 36. Harvey, P.W. and C. Sutcliffe, Adrenocortical hypertrophy: establishing cause and 31 toxicological significance. J. Appl. Toxicol., 2010. 30(7): p. 617-626. 32 37. Cohen, S., et al., Chronic stress, glucocorticoid receptor resistance, inflammation, and disease 33 risk. Proceedings of the National Academy of Sciences, 2012. 109(16): p. 5995-5999. 34 38. Kamijo, Y., et al., Fatal bilateral adrenal hemorrhage following acute toluene poisoning: a 35 case report. J. Toxicol. Clin. Toxicol., 1998. 36(4): p. 365-368. 36 39. Howard, M.O., et al., Suicide ideation and attempts among inhalant users: results from the 37 national epidemiologic survey on alcohol and related conditions. Suicide Life Threat. Behav., 38 2010. 40(3): p. 276-286. 39 40. McGirr, A., et al., Dysregulation of the sympathetic nervous system, hypothalamic–pituitary– 40 adrenal axis and executive function in individuals at risk for suicide. Journal of psychiatry & 41 neuroscience: JPN, 2010. 35(6): p. 399. 42 41. Melhem, N.M., et al., Blunted HPA axis activity in suicide attempters compared to those at 43 high risk for suicidal behavior. Neuropsychopharmacology, 2016. 41(6): p. 1447.

Page 24 of 48

Rose Crossin

1

2 Figure 1. Diagnosing adrenal insufficiency. Adrenal hypertrophy can be a result of stress or

3 adrenal insufficiency. Differentiation is based upon basal corticosterone levels, with further

4 localisation of adrenal insufficiency within the Hypothalamic-pituitary-adrenal (HPA) axis

5 based on adrenocorticotropic hormone (ACTH) and CRH levels (under basal or dynamic

6 testing conditions, which may include an insulin tolerance test (ITT)).

7

Page 25 of 48

Rose Crossin

A. Daily food intake B. Body weight

#### ** ** ## # ** 4 0 ## ## ** ## 4 0 0 ##

##

## ## ## A ir A ir 3 0 ## 3 0 0 ** CIT ** P a ir-fe d P a ir-fe d ** CIT 2 0 0 ** 2 0 Body weight (g)

Daily food (g/day) *

Exposure Abstinence 1 0 0 E x p o s u re A b s tin e n c e 1 0 20 30 40 50 60 70 80 90 20 30 40 50 60 70 80 90 PND PND

C. Spine length D. Rump width ** ** 2 2 ** # ** 5 0 ** 2 0 ** ##

1 8 A ir A ir * ** P a ir-fe d 4 0 P a ir-fe d 1 6 CIT CIT 1 4

Spine length (cm) 3 0

1 2 Rump width (mm)

1 0 Exposure Abstinence Exposure Abstinence 8 2 0 20 30 40 50 60 70 80 90 100 20 30 40 50 60 70 80 90 PND PND 1

2 Figure 2. Food intake and growth measurements during toluene exposure and

3 abstinence. A) Daily food intake was significantly reduced in the CIT group, and thus the

4 Pair-fed group from exposure day 3 onwards, but normalised to Air-control group during the

5 abstinence period. B) Body weight was decreased in the CIT group compared to both Air-

6 control and Pair-fed from exposure day 2 onwards. By the end of the exposure period, both

7 CIT and Pair-fed groups were significantly lower than Air-control. However, the Pair-fed

8 group normalised to Air-control during the abstinence period, whereas the CIT group

9 remained significantly lower at the end of abstinence. C) Spine length in the CIT group was

10 significantly lower than Air-control and Pair-fed from week 2 of exposure until the end of

11 abstinence, with the exception of one measurement at the end of exposure where the Pair-fed

12 group were also significantly lower than Air-control. D) At the end of exposure, both the CIT

13 and Pair-fed groups were significantly lower than Air, whereas at the end of abstinence, the

Page 26 of 48

Rose Crossin

1 CIT group was lower than both Air-control and Pair-fed groups. *p<0.05, **p<0.01 (CIT

2 significantly different from both Air-control and Pair-fed). #p<0.05, ##p<0.01 (CIT and Pair-

3 fed significantly different from Air-control). Measures taken throughout exposure and

4 abstinence periods (Air-control n=10, Pair-fed n=10, CIT n=12).

5

6

Page 27 of 48

Rose Crossin

A. V02 baseline - weight corrected B. Energy expenditure baseline - weight corrected

2 5 0 0 1 2 * (group effect) ** (group effect)

CIT 1 0 CIT 2 0 0 0 A ir A ir

8 m l.h .k g

1 5 0 0 k c a l.h .k g

6

1 0 0 0 M M M M M PM AM AM PM P PM AM AM A

A 0 0 0 0 0 0 0 0 P 0 0 0 0 P 0 0 0 :0 :0 :0 0 :0 : :0 0 :0 :0 0 : 0 0 : 0 0 0 0 0 0 0 0 0 0 0 :0 0 : :0 :0 0 : : :0 3 : 7 7 : 1 :0 0 3 : 7 3 1 :0 0 3 : 7 : 1 1 1 1 1 1 T im e T im e

C. Respiratory exchange ratio fast/re-feeding D. Food total baseline

1 .0 5 0 F a s tin g R e -fe e d in g

CIT 4 0 0 .9 A ir ** 3 0 0 .8 2 0

0 .7 1 0 Food Consumed (g)

Respiratory0 exchange .6 ratio 0 M M M A ir CIT P PM AM AM

A 0 0 0 0 0 P 0 0 0 : :0 0 :0 :0 0 0 : 0 0 0 0 0 :0 0 : : :0 7 3 1 :0 0 3 : 7 : 1 1 1

T im e

E. Food total re-feeding F. Drink re-feeding

1 5 2 5 * 2 0

1 0 1 5

1 0 5 mL consumed 5 Food Consumed (g)

0 0 A ir CIT A ir CIT 1

2 Figure 3. Calorimetric assessments reveal changes in energy expenditure in CIT rats. A)

3 CIT animals had increased baseline metabolic rate and (B) increased baseline energy

4 expenditure. C) CIT animals had increased respiratory exchange ratio in a re-feeding

5 paradigm following a fast. D.) No significant group differences were evident in baseline food

6 intake. E) No significant differences were evident in food intake in a re-feeding paradigm

Page 28 of 48

Rose Crossin

1 following a fast. F) CIT animals consumed significantly less water in a re-feeding paradigm

2 following a fast. *p<0.05, **p<0.01. Experiment undertaken at end of exposure period (Air-

3 control n=8, CIT n=8).

4

Page 29 of 48

Rose Crossin

A. Blood glucose - exposure B. Insulin - exposure

8 1 .5 ** A ir A ir

P a ir-fe d P a ir-fe d 7 *** ** CIT 1 .0 CIT

6 * 0 .5 5 Insulin (ng/mL) Blood glucose (mmol/L) 4 0 .0 S a te d F a s te d S a te d F a s te d

C. Liver glycogen - exposure D. Leptin - exposure

1 0 1 0 * ** A ir 8 8 P a ir-fe d

CIT 6 6

*** 4 4 * Leptin (ng/mL) 2 2 Liver glycogen (mg/mL) 0 0 A ir P a ir-fe d CIT S a te d F a s te d 1

2 Figure 4. Blood profiling shows metabolic disturbances in CIT rats. A) At the end of

3 exposure the CIT group had significantly reduced fasting blood glucose, compared to both

4 Air-control and Pair-fed groups. B) At the end of exposure, the Pair-fed group had

5 significantly lower sated insulin compared to the Air-control group, and in the fasted state the

6 CIT group had significantly lower insulin compared to the Pair-fed group. C). There were no

7 significant group differences in liver glycogen levels at the end of exposure. D) At the end of

8 exposure, both the Pair-fed and CIT groups had significantly lower leptin levels in both the

9 sated and fasted state, compared to the Air-control group. *p<0.05, **p<0.01, ***p<0.001.

10 Insulin, leptin and blood glucose were measured at the end of the exposure (Air-control n=20,

11 Pair-fed n=20, CIT n=23) and abstinence periods (Air-control n=10, Pair-fed n=10, CIT

12 n=12). Glycogen was tested in liver collected post-mortem at the end of the exposure (Air-

13 control n=10, Pair-fed n=10, CIT n=11) and abstinence periods (Air-control n=10, Pair-fed

14 n=10, CIT n=12).

Page 30 of 48

Rose Crossin

A. Trabecular bone volume B. Trabecular thickness

1 5 E xp o su re A b s tin e n c e 8 0 E xp o su re A b s tin e n c e **

1 0 7 0

5 6 0

0 Trabecular5 0thickness (µm) Trabecular bone volume (%) Air Pair-fed CIT Air Pair-fed CIT Air Pair-fed CIT Air Pair-fed CIT

C. Trabecular number D. Trabecular separation E xp o su re A b s tin e n c e 2 .0 1 5 0 0 E xp o su re A b s tin e n c e ** **

1 .5 1 0 0 0

1 .0

5 0 0 0 .5 Trabecular number (1/mm)

0 .0 Trabecular separation0 (µm) Air Pair-fed CIT Air Pair-fed CIT Air Pair-fed CIT Air Pair-fed CIT

E. Cortical thickness F. Tissue Mineral Density

7 5 0 E xp o su re A b s tin e n c e 1 .5 E xp o su re A b s tin e n c e

7 0 0 **

1 .0 6 5 0

6 0 0 0 .5 TMD (g/cm3) 5 5 0 Cortical thickness (µm) 5 0 0 0 .0 Air Pairfed CIT Air Pair-fed CIT Air Pairfed CIT Air Pair-fed CIT 1

2 Figure 5. Skeletal structural differences revealed in CIT rats. A) Trabecular bone volume

3 was significantly lower in the Pair-fed group compared to both Air-control and CIT at the end

4 of exposure, with no group differences at the end of abstinence. B) There were no significant

5 group differences in trabecular thickness at either time point. C) Trabecular number was

6 significantly lower in the Pair-fed group compared to both Air-control and CIT at the end of

7 exposure, with no group differences at the end of abstinence. D) Trabecular separation was

8 significantly greater in the Pair-fed group compared to both Air-control and CIT at the end of

9 exposure, with no group differences at the end of abstinence. E) There were no significant

10 group differences in cortical thickness at the end of exposure, though at the end of abstinence

11 the CIT group was significantly lower than both the Air-control and Pair-fed groups. F) There

12 were no group differences in tissue mineral density at either time point. *p<0.05, ** p<0.01.

Page 31 of 48

Rose Crossin

1 Tibiae were analysed by µCT at the end of the exposure (Air-control n=10, Pair-fed n=10,

2 CIT n=11) and abstinence periods (Air-control n=10, Pair-fed n=10, CIT n=12).

3

Page 32 of 48

Rose Crossin

A. Zona fasiculata - width B. Zona fasiculata - cell size

1 .0 E xp o su re A b s tin e n c e 2 5 E xp o su re A b s tin e n c e * **** **** **** 0 .8 2 0

0 .6 1 5

0 .4 1 0

Zone0 width .2 (mm) 5

0 .0 Average cell0 size (microns) Air CIT Air CIT Air CIT Air CIT

C. Corticosterone - end exposure D. Corticosterone - end abstinence

A ir 1 2 0 1 2 0 * A ir CIT 1 0 0 1 0 0 CIT

8 0 8 0

6 0 6 0

4 0 4 0

2 0 2 0 Corticosterone ng/mL Corticosterone ng/mL 0 0 S a te d F a s te d S a te d F a s te d

E. Basal ACTH - end exposure

2 .5 *

2 .0

1 .5

1 .0 ACTH (ng/mL) 0 .5

0 .0 A ir CIT 1 2

3 Figure 6. Adrenal histology and blood profiling highlight adrenal differences in CIT

4 rats. A) CIT animals had significantly larger zona fasiculata width compared to Air-control,

5 at the end of both exposure and abstinence. B) CIT animals had significantly larger average

6 cell sizes within the zona fasiculata compared to Air-control, at the end of both exposure and

Page 33 of 48

Rose Crossin

1 abstinence. * p<0.05, ****p<0.0001. Adrenal glands collected at the end of the exposure

2 (Air-control n=10, CIT n=11) and abstinence periods (Air-control n=10, CIT n=12). C) No

3 significant group differences were observed in basal corticosterone at the end of exposure in

4 either sated or fasted states. D) CIT animals had significantly lower basal corticosterone

5 levels compared to Air-control, in the sated state, at the end of abstinence. E) Basal ACTH

6 was increased in the CIT group at the end of the exposure period. * p<0.05. Corticosterone:

7 end of exposure (Air-control n=20, CIT n=23) and abstinence (Air-control n=10, CIT n=12).

8 ACTH: end of exposure (Air-control n=8, CIT n=9).

9

Page 34 of 48

Rose Crossin

A. Insulin tolerance test - blood glucose B. Insulin tolerance test - corticosterone

8 3 0 0 *** ** ** ** * 6 2 0 0 CIT A ir 4 CIT 1 0 0 A ir 2 Blood glucose (mmol/L) Corticosterone (ng/mL) 0 0 Baseline15 30 45 60 90 B a s e lin e 3 0 6 0 9 0

T im e T im e

C. Stress response test - ACTH D. Stress response test - corticosterone

3 5 0 0 CIT CIT 4 0 0 * A ir A ir 2 3 0 0

2 0 0 1 * ACTH (ng/mL) 1 0 0 Corticosterone (ng/mL) 0 0 Baseline 0 30 60 90 Baseline 0 30 60 90

T im e T im e

E. Stress response test - blood glucose

1 1 ** CIT

1 0 A ir

9

8

7 Blood glucose (mmol/L) 6 Baseline0 15 30 45 60 90 T im e 1

2 Figure 7. Dynamic testing of the HPA axis revealed differences in CIT rats. A) CIT

3 animals had lower blood glucose levels in response to an ITT, between 30-60 minutes post

4 insulin administration. B) CIT animals had higher corticosterone levels in response to an ITT

5 between 30-60 minutes post insulin administration. * p<0.05, ** p<0.01, ***p<0.001. Group

Page 35 of 48

Rose Crossin

1 numbers at end of exposure (Air-control n=8, CIT n=9). C) CIT animals showed greater basal

2 ACTH but impairment in the change from baseline to time 0. D) CIT animals showed

3 elevated corticosterone levels at the end of the stress response test. E) CIT animals showed

4 decreased blood glucose response at time 0 in the stress response test. * p<0.05, ** p<0.01.

5 Group numbers at end of exposure (Air-control n=8, CIT n=9).

6

Page 36 of 48

Rose Crossin

1

2 Figure 8. CIT rats show a blunted blood glucose response to changing corticosterone

3 levels. The blood glucose response to changing corticosterone levels, within the context of a

4 stress response test, was blunted in the CIT group compared to Air-control, with twice the

5 amount of change to corticosterone needed in the CIT group, to achieve the same change in

6 blood glucose.

7

Page 37 of 48

Rose Crossin

1 Table 1. Post-mortem organ weights after exposure and after abstinence for CIT and Air 2 animals.

Organ End of exposure organ weight (g) End of abstinence organ weight (g) (organ weight / g body weight) (organ weight / g body weight) Air CIT Air CIT 1.82±0.02 1.78±0.02 1.90±0.02 1.88±0.01 Brain (0.00557± (0.00545± (0.00405± (0.00428± 5.70063E-05) 6.0347E-05) 0.000084) 0.000084) 1.18±0.06 1.19±0.05 1.35±0.08 1.41±0.06 Heart (0.00360± (0.00364± (0.00287± (0.00319± 0.000194) 0.000147) 0.000174) 0.000125) 2.26±0.21 1.66±0.11 1.91±0.19 1.89±0.14 Lung (0.00690± (0.00509± (0.00407± (0.00427± 0.000656) 0.000347) 0.000408) 0.000281) 14.53±0.36 12.66±0.76 17.07±0.63 17.48±0.54 Liver (0.04434± (0.03864± (0.03626± (0.03961± 0.001084) 0.002314) 0.000925) 0.000643) *** 1.27±0.02 1.09±0.04 1.58±0.05 1.46±0.05 Kidney (0.00388± (0.00332± (0.00336± (0.00332± 4.87E-05) 0.000118) 0.000095) 0.000071) 0.027±0.001 0.030±0.002 0.031±0.002 0.033±0.001 Adrenal (0.00008± (0.00009± (6.5493E-05± (7.5853-05± 3.4E-06) 4.83E-06) ** 0.000004) 0.000002) 1.76±0.09 1.48±0.08 3.43±0.25 3.15±0.18 Fat pad (0.00537± (0.00452± (0.00728± (0.00711± 0.000289) 0.000245) 0.000487) 0.000329) 1.60±0.03 1.48±0.04 1.88±0.04 1.93±0.05 Teste (0.00488± (0.00452± (0.00401± (0.00440± 0.000105) 0.000118) 0.000103) 0.000127) 3

4 Adrenal glands were heavier in the CIT group, when corrected for body weight at end of

5 exposure. At end of abstinence, livers were heavier in the CIT group, when corrected for

6 body weight. Fat pad weight was the epididymal fat pad. ** p<0.01, ***p<0.001, based on

7 ANCOVA in which body weight was the co-variate. Organ weights were from the end of the

8 exposure (Air-control n=10, Pair-fed n=10, CIT n=11) and abstinence periods (Air-control

9 n=10, Pair-fed n=10, CIT n=12).

10

Page 38 of 48

Rose Crossin

1 Supplementary Information

2 Supplementary file 1 – pair-feeding protocol

3 The food allocation for the pair-fed group was based on the average consumption of the CIT

4 group from the previous monitoring period + a predicted increase in food consumption (as

5 rats were in a growth phase) ± a correction to ensure that the cumulative food intake

6 remained equal between the CIT and pair-fed groups.

7 Supplementary file 2 – Bone mechanical testing – 3 point bend

8 Each tibia was rehydrated overnight in phosphate buffered saline (PBS) at room temperature

9 prior to testing. To determine the mechanical properties of cortical bone each tibia was

10 loaded to failure at 0.5 mm/s using a Bose Biodynamic 5500 Test Instrument (Bose, DE,

11 USA). The span between the lower supports was 10 mm. Prior to testing, the tibiae were kept

12 moist in gauze swabs soaked in PBS. Bones were positioned such that the load was applied

13 8.75 mm from the top of distal condyle in the anterior-posterior (AP) direction with distal

14 condyle facing downwards. WinTest software (WinTest 7) was used to collect the load-

15 displacement data across 10 s with a sampling rate of 250 Hz. Structural properties including

16 Ultimate force (FU; N), yield force (FY; N), stiffness (S; N/mm), and energy (work) to

17 failure (U; mJ) endured by the tibia were calculated from the load and displacement data. The

18 yield point was determined from the load displacement curve at the point at which the curve

19 deviated from linear. Widths of the cortical mid-shaft in the medio-lateral (ML) and antero-

20 posterior (AP) directions, moment of inertia (Imin), and the average cortical thickness were

21 determined byμCT in the cortical region. Tibial material properties, i.e., stress-strain curves

22 were calculated from the structural properties (i.e., load-displacement curve) in combination

23 with morphological data from μCT. The obtained stress-strain curves reflect the stiffness,

24 strength and failure properties of the bone material itself without the influence of geometry.

Page 39 of 48

Rose Crossin

1 Supplementary file 3 – Statistical tests

2 Aim 1 – what is the metabolic and energy balance phenotype arising from inhalant

3 abuse and does it persist into abstinence?

4 All food intake, water intake and growth measures were analysed as a RM ANOVA (time

5 and group) with Tukey’s post-hoc testing. Bomb calorimetry results were analysed using a

6 one-way ANOVA with Tukey’s post-hoc at the end of both exposure and abstinence. 3-bottle

7 choice was analysed using a two-way ANOVA (liquid and group) with Tukey’s post-hoc

8 testing. Metabolic cage repeated measures data were analysed as a RM ANOVA (time and

9 group) with Tukey’s post-hoc testing. Food and water totals consumed within the metabolic

10 cages were analysed with independent samples t-tests. Blood glucose, insulin, and leptin

11 levels were analysed as a two-way ANOVA (fed-state and group) with Tukey’s post-hoc.

12 Liver glycogen levels were analysed as a one-way ANOVA (by group) with Tukey’s post-

13 hoc. All skeletal variables were measured as a one-way ANOVA (by group) with Tukey’s

14 post-hoc.

15 Aim 2 – is adrenal insufficiency present following adolescent inhalant abuse?

16 Post-mortem organ weights were analysed with ANCOVA, with body weight as the co-

17 variate. Adrenal histology results were analysed with independent sample t-tests. Basal

18 corticosterone and ACTH results were analysed with independent sample t-tests. Variables

19 within the ITT and stress response test were all measured as a two-way ANOVA (time and

20 group).

21 Supplementary file 4 – additional food and water intake results

22 There were no significant group differences in food intake when corrected for body weight,

23 during either exposure or abstinence (Supplementary Figure 1A). Daily water intake was

Page 40 of 48

Rose Crossin

1 increased in the CIT group compared to both Air-controls and Pair-fed during the exposure

2 period (F=5.465 p=0.01, Supplementary Figure 1B) with no group differences at the end of

3 the abstinence period. When corrected for body weight, water intake was again significantly

4 higher in the CIT group in the exposure period (F=16.946 p<0.000, Supplementary Figure

5 1C) which remained significant into the abstinence period (F=6.934 p=0.003, Supplementary

6 Figure 1C). There were no significant group differences in the conversion of food to body

7 weight during the exposure period, though the Pair-fed had a significantly higher conversion

8 of food to body weight during the abstinence period (F=4.790 p=0.016, Supplementary

9 Figure 1D).

A. Daily food intake by body weight B. Daily water intake

0 .2 5

* 6 0 0 .2 0 *

* A ir A ir 0 .1 5 4 0 P a ir-fe d P a ir-fe d

CIT CIT 0 .1 0

Water2 (mL/day) 0

Food0 (g/g .0 body5 weight)

E x p o s u re A b s tin e n c e E x p o s u re A b s tin e n c e 0 .0 0 0 20 30 40 50 60 70 80 90 20 30 40 50 60 70 80 90 PND PND

C. Water intake corrected for weight D. Food utilisation ratio

0 .3 *** ****** 3 *** *** 2

0 .2 *** A ir 1 A ir *** *** P a ir-fe d P a ir-fe d ## 0 .3 5 CIT ## CIT

## 0 .1 0 .3 0 ## ##

Water (mL/g body weight) 0 .2 5 E x p o s u re A b s tin e n c e Exposure Abstinence 0 .0 Body0 weight .2 (g)0 / cumulative food (g) 20 30 40 50 60 70 80 90 20 30 40 50 60 70 80 90 PND PND 10

11 Supplementary Figure 1. A) There were no significant group differences in food intake when 12 corrected for body weight. B) Daily water intake was higher in the CIT group compared to 13 both Air-controls and Pair-fed during the exposure, but not abstinence period. C) Daily water 14 intake when corrected for body weight was higher in the CIT group compared to Air-controls

Page 41 of 48

Rose Crossin

1 and Pair-fed through exposure and abstinence D) There were no group differences in the 2 conversion of food to body weight during the exposure period, but the Pair-fed group was 3 significantly higher than Air-controls and CIT during the abstinence period. *p<0.05, 4 ***p<0.001 (CIT significantly different from both Air-controls and Pair-fed). ##p<0.01 5 (Pair-fed significantly different from Air-controls and CIT). Throughout exposure and 6 abstinence periods (Air-controls n=10, Pair-fed n=10, CIT n=12).

7

8 Supplementary file 5 – additional metabolic cage results

9 There were no significant differences in metabolic rate when body weight was not corrected

10 for, in either baseline (Supplementary Figure 2A) or in a fast/re-feeding paradigm

11 (Supplementary Figure 2B). There was also no significant difference in metabolic rate when

12 body weight was corrected for, in a fast/re-feeding paradigm (Supplementary Figure 2C).

13 Similarly, there were no significant differences in energy expenditure when body weight was

14 not corrected for, in either baseline (Supplementary Figure 2D) or in a fast/re-feeding

15 paradigm (Supplementary Figure 2E), or when body weight was corrected for, in a fast/re-

16 feeding paradigm (Supplementary Figure 2F). There were no significant differences in RER

17 under baseline conditions (Supplementary Figure 2G) or water intake under baseline

18 conditions (Supplementary Figure 2H).

Page 42 of 48

Rose Crossin

A. V02 baseline B. V02 fast/re-feeding

7 0 0 7 0 0 F a s tin g R e -fe e d in g

CIT CIT 6 0 0 6 0 0 A ir A ir

5 0 0 5 0 0 m l.h m l.h

4 0 0 4 0 0

3 0 0 3 0 0 M M M M M M P PM AM AM P PM AM AM

A

A 0 0 0 0 0 0 0 0 P 0 0 0 0 0 P 0 0 0 : :0 0 :0 :0 : :0 0 :0 :0 0 0 : 0 0 : 0 0 0 0 0 0 0 0 0 0 :0 0 : : :0 :0 0 : : :0 7 3 1 :0 0 3 : 7 : 7 3 1 :0 0 3 : 7 : 1 1 1 1 1 1

T im e T im e

C. V02 fast/re-feeding - weight corrected D. Energy expenditure baseline

2 5 0 0 3 .5 Fasting Re-feeding CIT CIT 3 .0 2 0 0 0 A ir A ir

2 .5 k c a l.h m l.h .k g 1 5 0 0 2 .0

1 0 0 0 M M M M M M P P PM AM AM AM AM PM

A A 0 0 0 0 0 0 0 0 0 P 0 0 0 P 0 0 0 0 : : :0 0 :0 :0 :0 :0 0 :0 0 0 0 : 0 : 0 0 0 0 0 0 0 0 0 0 : :0 0 : : :0 0 : :0 :0 3 7 3 7 3 : 7 : 1 :0 0 7 : 1 :0 0 3 : 1 1 1 1 1 1 T im e T im e

E. Energy expenditure fast/re-feeding F. Energy expenditure fast/re-feeding - weight corrected

3 .5 1 2 F a s tin g R e -fe e d in g F a s tin g R e -fe e d in g

CIT CIT 3 .0 A ir 1 0 A ir

2 .5 8 k c a l.h k c a l.h .k g 2 .0 6 M M M M M M M P P PM PM AM AM AM A

A

A 0 0 0 0 0 0 0 0 0 0 P 0 0 0 P 0 0 0 0 0 : : :0 0 :0 :0 :0 :0 : 0 0 0 : 0 0 : 0 0 0 0 0 0 0 0 0 0 0 : :0 0 : :0 0 : : :0 : :0 3 7 7 3 1 :0 0 1 : 3 : 3 : 7 : 7 1 1 1 1 1 1

T im e T im e

G. Respiratory exchange ratio baseline H. Drink total baseline 1 .0 1 5 0

0 .9

0 .8 1 0 0

CIT 0 .7 A ir

0 .6 5 0 M mL consumed PM PM AM AM A 0 0 0 0 :0 :0 0 :0 0 :0 0 : 0 0 :0 0 P M 0 0 0 0 :0 7 : 3 : 1 :0 0 3 :0 0 7 : 0 1 1 1 A ir CIT T im e 1

2 Supplementary Figure 2. A) No significant group differences were evident in baseline 3 metabolic rate when body weight is not corrected for. B) No significant group differences 4 were evident in metabolic rate when body weight is not corrected for, in a fast/re-feeding

Page 43 of 48

Rose Crossin

1 paradigm. C) No significant group differences were evident in metabolic rate when body 2 weight is corrected for, in a fast/re-feeding paradigm. D) No significant group differences 3 were evident in baseline energy expenditure when body weight is not corrected for. E) No 4 significant group differences were evident in energy expenditure when body weight is not 5 corrected for, in a fast/re-feeding paradigm. F) No significant group differences were evident 6 in energy expenditure when body weight is corrected for, in a fast/re-feeding paradigm. G) 7 No significant group differences in baseline respiratory exchange ratio. H) No significant 8 group differences were evident in baseline water intake. Experiment undertaken at end of 9 exposure period (Air-controls n=8, CIT n=8).

10 Supplementary file 6 – additional skeletal data

11 No significant differences at the end of exposure or abstinence periods were observed by

12 microCT for tibiae length (Supplementary Figure 3A), marrow area (Supplementary Figure

13 3B), cortical area (Supplementary Figure 3C), endocortical perimeter (Supplementary Figure

14 3D), periosteal perimeter (Supplementary Figure 3E), or mean polar moment of inertia

15 (Supplementary Figure 3F).

16 As there had been an observed reduction in cortical thickness in the CIT group only at the

17 end of the abstinence period, the mechanical properties of the bone were tested at that time

18 point. There were no significant differences in material properties of the tibiae at the end of

19 abstinence for variables of Young’s Modulus (Supplementary Figure 4A), Yield Stress

20 (Supplementary Figure 4B), Ultimate Stress (Supplementary Figure 4C) or Failure Stress

21 (Supplementary Figure 4D).

Page 44 of 48

Rose Crossin

A. Tibiae Length B. Marrow area E xp o su re A b s tin e n c e 4 0 E xp o su re A b s tin e n c e 6

3 0 4

2 0 m m m m 2 2 1 0

0 0 Air Pair-fed CIT Air Pair-fed CIT Air Pairfed CIT Air Pair-fed CIT

C. Cortical area D. Endocortical perimeter

8 E xp o su re A b s tin e n c e 1 5 E xp o su re A b s tin e n c e

6 1 0

4 m m m m 2 5 2

0 0 Air Pairfed CIT Air Pair-fed CIT Air Pairfed CIT Air Pair-fed CIT

E. Periosteal perimeter F. Mean polar moment of inertia

2 0 E xp o su re A b s tin e n c e 2 5 E xp o su re A b s tin e n c e

2 0 1 5

1 5 1 0 m m m m 4 1 0

5 5

0 0 Air Pairfed CIT Air Pair-fed CIT Air Pairfed CIT Air Pair-fed CIT 1

2 Supplementary Figure 3. No significant group differences were observed at the end of 3 exposure or abstinence for A) tibiae length. B) marrow area. C) cortical area. D) endocortical 4 perimeter. E) periosteal perimeter. F) mean polar moment or inertia. Tibiae were analysed by

Page 45 of 48

Rose Crossin

1 µCT at the end of the exposure (Air-controls n=10, Pair-fed n=10, CIT n=11) and abstinence 2 periods (Air-controls n=10, Pair-fed n=10, CIT n=12).

A. Youngs Modulus B. Yield stress

5 0 0 1 5

4 0 0

1 0 3 0 0 M P a 2 0 0 M P a 5

1 0 0

0 0 A ir P a ir-fe d CIT A ir P a ir-fe d CIT

C. Ultimate stress D. Failure stress

2 0 2 0

1 5 1 5

1 0 1 0 M P a M P a

5 5

0 0 A ir P a ir-fe d CIT A ir P a ir-fe d CIT 3

4 Supplementary Figure 4. No significant group differences were observed at the end of 5 abstinence for A) Young’s Modulus. B) Yield Stress. C) Ultimate Stress. D) Failure Stress. 6 Tibiae were mechanically tested at the end of the abstinence period (Air-controls n=10, Pair- 7 fed n=10, CIT n=12).

8 Supplementary file 7 – additional adrenal histology data

9 In the medulla, the CIT group had a significantly decreased zone width at the end of exposure

10 (t=5.69 p<0.0001, Supplementary Figure 5A), with no significant differences at the end of

11 abstinence, however, the CIT group had a significantly increased cell size in the medulla at

12 the end of exposure (t=3.361 p=0.004, Supplementary Figure 5B), with no differences at the

13 end of abstinence. In the zona glomerulosa there were no group differences in zone width at

14 either time point (Supplementary Figure 5C), however, the CIT group had significantly

15 smaller cell size at the end of exposure (t=2.296 p=0.0339, Supplementary Figure 5D) and

Page 46 of 48

Rose Crossin

1 abstinence (t=2.618 p=0.0165, Supplementary Figure 5D). In the zona reticularis there were

2 no group differences in zone width at either time point (Supplementary Figure 5E), however,

3 the CIT group had significantly smaller cell size at the end of exposure (t=2.221 p=0.0394,

4 Supplementary Figure 5F) and abstinence (t=2.264 p=0.0349, Supplementary Figure 5F).

A. Medulla width B. Medulla cell size

2 .0 E xp o su re A b s tin e n c e 2 5 E xp o su re A b s tin e n c e

**** 2 0 ** 1 .5

1 5 1 .0 1 0

0 .5

Zone width (mm) 5

0 .0 Average cell0 size (microns) Air CIT Air CIT Air CIT Air CIT

C. Zona glomerulosa width D. Zona glomerulosa cell size

0 .1 5 E xp o su re A b s tin e n c e 1 5 E xp o su re A b s tin e n c e

* *

0 .1 0 1 0

0 .0 5 5 Zone width (mm)

0 .0 0 Average cell0 size (microns) Air CIT Air CIT Air CIT Air CIT

E. Zona reticularis width F. Zona reticularis cell size

0 .6 E xp o su re A b s tin e n c e 1 5 E xp o su re A b s tin e n c e

* *

0 .4 1 0

0 .2 5 Zone width (mm)

0 .0 Average cell0 size (microns) Air CIT Air CIT Air CIT Air CIT 5 6 Supplementary Figure 5. A) CIT group had a reduced medulla zone width at the end of

Page 47 of 48

Rose Crossin

1 exposure, but no difference at the end of abstinence. B) CIT group had increased medulla cell 2 size at the end of exposure, but no difference at the end of abstinence. C). There were no 3 group differences in zona glomerulosa width at either time point. D). CIT group had 4 decreased zona glomerulosa cell size at both time points. E). There were no group differences 5 in zona reticularis width at either time point. F) CIT group had decreased zona reticularis cell 6 size at both time points. * p<0.05, **p<0.01, ****p<0.0001. Adrenal glands collected at the 7 end of the exposure (Air-controls n=10, CIT n=11) and abstinence periods (Air-controls 8 n=10, CIT n=12).

9

10

Page 48 of 48

Rose Crossin (737900)

Supplementary Data for Chapter 4

Prelude This section describes findings related to characterising the mechanisms underlying the growth and energy balance consequences of inhalant abuse (Chapter 4) that either failed methodologically, or did not result in data considered necessary for inclusion in the narrative of the manuscript. However, they are included here to illustrate that attempts were made to fully characterise the effects of inhalant exposure on the energy balance equation. As part of aim 1 (characterise the effects of adolescent inhalant abuse using the energy balance equation, and to determine the persistence of these effects into sustained abstinence), the energy content of faecal samples were assessed in order to identify potential changes to nutrient absorption. In addition to this, assessment of the faecal microbiome was also to be conducted. This had never been conducted in a toluene exposure or inhalant abuse model, and would be used to identify whether the gut should be a key organ of interest for future studies, particularly relevant given that inhalant users frequently report gastrointestinal symptoms. Also in relation to aim 1, it was intended to delineate energy expenditure into its components by specifically measuring physical activity and whole body thermogenesis. Doing so would enable a deeper understanding of energy expenditure changes following inhalant abuse. As part of the characterisation studies, organ weights had been measured at the end of both exposure and abstinence periods. Adrenal hypertrophy was evident at the end of exposure, which led to a more detailed investigation of that organ, as described in the manuscript. However, liver hypertrophy was also identified at the end of abstinence period, and to investigate potential causes of this, liver lipids were to be quantified to determine if fatty liver disease was present and could potentially account for the observed hypertrophy.

In relation to aim 2 (identify a mechanism underlying the observed growth effects), after under-nutrition had been rejected as the sole mediator of inhalant-induced growth changes, the hypothesis that adolescent inhalant abuse may result in adrenal insufficiency and thus underlie the growth effects was tested. Adrenal insufficiency is characterised by a lack of the hormone cortisol (corticosterone in rats), which is a hormone with anti-inflammatory effects. Therefore, it was hypothesised that adrenal insufficiency should result in a pro-inflammatory state, and as a preliminary step before a full cytokine array be undertaken, levels of Interleukin-1 beta (IL-1β, a cytokine associated with anorexic effects) were tested in the liver and hypothalamus. These are key metabolic organs, and inflammation in these organs may lead to a disruption of metabolic and energy balance processes. Additionally, if adrenal insufficiency was present in early adolescence, this had the potential to result in delayed skeletal maturation and thus a reduction in bone age (relative to actual age), therefore, an assessment of changes to bone age may have been able to provide evidence to support the adrenal insufficiency hypothesis. Lastly, adrenal insufficiency is treated clinically with cortisol replacement. As part of the diagnostic process for adrenal insufficiency, it was aimed to determine if the energy balance changes arising from adolescent inhalant abuse could either be prevented from occurring (by corticosterone replacement during the exposure period) or corrected during abstinence (by corticosterone replacement during the abstinence period). This experiment would form part of the identification of adrenal insufficiency, but also identify

120 Rose Crossin (737900) whether cortisol treatment may be a useful therapy to consider for inhalant users who had growth and energy balance impairments.

Methods

Faecal microbiome Fresh faeces were collected from cages of individually housed animals at the end of both the exposure and abstinence periods and frozen at -80°C. As this was a pilot study, faeces from 6 animals per group (Air-control, CIT) were randomly selected for this experiment, with the same animals used at both time points to provide a repeated measure. Approximately 0.25 g faeces / animal were extracted while frozen and provided to an external collaborator for preparation and analysis of the faecal microbiome using 16S rRNA gene sequencing.

Physical activity Indirect calorimetry was utilised at the end of the exposure period (Air-control n=8, CIT n=8) to measure oxygen consumption and carbon dioxide production. Animals were housed within enclosed calorimetry chambers (LabMaster; TSE-systems, Bad Homburg, Germany) for 48 hours (baseline), followed by an 18 hour fast (water was available ad libitum), then a 6 hour re- feeding period. This system should also provide a measure of physical activity, using a sensor that measures movement from the centre of the cage, which can then be used to quantify activity levels.

Thermographic imaging Thermographic imaging was conducted at the end of exposure period, following indirect calorimetry (Air-control n=8, CIT n=8). This method was trialled as it was non-invasive and would allow for repeated measures to be taken in a single animal. Thermographic imaging was performed by first clipping the fur over the BAT and the lumbar spine (to serve as a control region). Animals were returned to their home cages for 30 minutes after clipping to minimise stress activation. Each animal was then placed in an open box, with an infrared camera (FLIR T650sc, temperature range set to 30°C) mounted 100 cm directly above. Animals were rested in the open box for 5 minutes before taking the first image. Multiple images were taken of each animal, discarding those where the animal was in a rearing or hunched position. Images were analysed using camera-specific software (ResearchIR 4.0), with 240 pixel circular field of interest positioned over the BAT and lumbar region. The mean and maximum temperatures were quantified for each region. Group differences were assessed using unpaired t-tests.

Liver lipids Livers were collected post-mortem at the end of exposure (Air-control n=10, CIT n=11) and abstinence (Air-control n=10, CIT n=12) periods, snap frozen in liquid nitrogen and then stored at -80°C until needed. Samples of frozen liver were mounted using Tissue-Tek® before being sectioned on the cryostat at 16°C. 12 µm slices were taken, at three different depths (with slices taken in triplicate at each depth), and mounted on gelatinised slides, before storage at - 80°C. Two Oil Red O staining methods were trialled to visualise lipids. The first used an Oil Red O staining kit (Abcam scientific) that had staining solutions pre-made, and the second solution prepared from powdered Oil Red O. Solution was prepared with 0.5 g Oil Red O in 100 mL isopropyl alcohol that was stood overnight, then filtered, before being made into working solution with 60 mL stock Oil Red O solution into 40 mL dextrin solution (1g dextrin in 100 mL

121 Rose Crossin (737900) distilled water). Both methods were trialled both with and without an H&E counter-stain. Incubation and staining protocol was conducted as per [154]. Images were captured using a light microscope at x20 magnification.

IL-1β IL-1β was measured in the hypothalamus and liver, from organs collected at the end of both exposure (Air-control n=10, CIT n=11) and abstinence (Air-control n=10, CIT n=12), that were snap frozen in liquid nitrogen and then stored at -80°C. Liver supernatant was prepared from samples collected at post-mortem, in line with ELISA kit instructions. The hypothalamus was dissected from brains: 2 x 600 µm slices were taken, beginning at Bregma -1.56. The hypothalamus was collected by taking 2 x 2 mm punches from each slice. Hypothalamus supernatant was then prepared from this punched tissue. Levels of IL-1β were measured by ELISA kit (abcam ab100768). Group differences were assessed using unpaired t-tests.

Assessment of bone age via skeletal µCT Images of tibiae collected at the end of exposure (Air-control n=10, CIT n=11) and abstinence (Air-control n=10, CIT n=12) were acquired as described in Chapter 4. Growth plate thickness was used as a measure of bone age. In CTAn a region of interest (1 mm x 1 mm) was positioned over the growth plate to acquire a new image of the growth plate; the boundaries of which were determined first by classifying a region of interest based upon thresholding levels, and second by hand-tracing the region of interest in two dimensions, to isolate the growth plate from the surrounding trabecular bone and create a 3-dimensional image of the growth plate. Structure thickness and density were assessed with CTAn.

Corticosterone treatment This experiment included an additional CIT group that were treated with corticosterone at 25 mg/L in drinking water [155] throughout the 4-week toluene exposure protocol (CIT-CE, corticosterone during exposure). This experiment utilised a cross-over design, such that animals in the CIT group were then treated with corticosterone during the 4-week abstinence period (CIT-CA, corticosterone during abstinence). Importantly, this experimental design meant that both CIT groups received corticosterone treatment at some point during the protocol. Group sizes were (Air-control n=8), CIT-CA (n=9), CIT-CE (n=9). Energy balance (food intake and energy expenditure) and growth variables (body weight and spine length) were assessed as described in Chapter 4. Group differences were analysed using RM-ANOVA.

Statistical methods Statistical analysis was performed using SPSS (IBM Version 22). Repeated measures were analysed as a two-way ANOVA over time. Comparisons between single measures for two groups were analysed with an independent samples t-test. Results are reported as mean±standard error of the mean (SEM) and considered significant at p<0.05.

Results

Faecal microbiome At the time of writing, this analysis is still to be completed by the collaborator, and thus, no results can be reported.

122 Rose Crossin (737900)

Physical activity At the end of the 3 day study protocol within the calorimetry chambers it was identified that there had been a centralised software failure, which meant that none of the activity sensors had recorded any activity. Due to equipment and experimental constraints, this experiment could not be repeated, and thus, no results can be reported.

Thermographic imaging There were no significant group differences for mean or maximum temperature in either the BAT or lumbar regions (all p values >0.05, Figure 5).

B A T m e a n te m p B A T m a x te m p

3 8 .0 3 8 .0

3 7 .5 3 7 .5

C

C

s

s

e e

e 3 7 .0 e

3 7 .0 r

r

g

g

e

e

d d 3 6 .5 3 6 .5

3 6 .0 3 6 .0 A ir C IT A ir C IT

L u m b a r m e a n te m p L u m b a r m a x te m p

3 8 .0 3 8 .0

3 7 .5 3 7 .5

C C

s s

e e

e 3 7 .0 e 3 7 .0

r r

g g

e e

d d 3 6 .5 3 6 .5

3 6 .0 3 6 .0 A ir C IT A ir C IT

Figure 5 – Whole body thermographic imaging. No significant differences were identified for mean or maximum temperature in either the BAT or lumbar regions, using whole body thermography.

123 Rose Crossin (737900)

Liver lipids None of the trialled staining methods were successful, with no lipids visible in any sample. Experiments were repeated multiple times, in consultation with the histology service at the Florey, but lipid staining was not successful, and thus, no results are reported.

IL-1β There were no significant group differences between IL-1β levels in either the liver or hypothalamus, at either time point (all p values >0.05, Figure 6). However, there was a trend towards increased IL-1β in the hypothalamus at the end of the abstinence period in the CIT group (p=0.064) and thus further exploration of inflammatory markers in the CIT model would be beneficial.

IL -1 b e ta L iv e r e n d e x p o s u r e IL -1 b e ta L iv e r e n d a b s tin e n c e

3 0 0 0 3 0 0 0

2 0 0 0 2 0 0 0

L

L

m

m

/

/

g

g

p p 1 0 0 0 1 0 0 0

0 0 A ir C IT A ir C IT

IL -1 b e ta h y p o th a la m u s e n d e x p o s u r e IL -1 b e ta h y p o th a la m u s e n d a b s tin e n c e

1 5 0 0 1 5 0 0

p = 0 .0 6 4

1 0 0 0 1 0 0 0

L

L

m

m

/

/

g

g

p p 5 0 0 5 0 0

0 0 A ir C IT A ir C IT

Figure 6 – IL-1β in the liver and hypothalamus. No significant group differences were observed in either the liver or hypothalamus at the end of exposure and abstinence. It was noted however that there was a trend towards increased IL-1β in the hypothalamus at the end of the abstinence period in the CIT group and thus further exploration of inflammatory markers in the CIT model would be beneficial.

Assessment of bone age via skeletal µCT Neither method was able to correctly isolate the growth plate; upon review of the images and the data for structure thickness and density it was evident that the growth plate boundaries were too indistinct in the 3-dimensional image for the software to be able to correctly isolate the region of interest. Therefore, the growth plate images included trabecular bone. Various

124 Rose Crossin (737900) thresholding levels were trialled, upon advice from the software manufacturer, but none were successful, and thus, no results are reported.

Corticosterone treatment Corticosterone treatment during both exposure and abstinence was unable to differentiate the two CIT groups on any of the energy balance variables. While CIT-CE and CIT-CA groups showed significantly reduced body weight, food intake, and spine length, and increased energy expenditure compared to Air-controls, there were no differences between the two CIT groups (all p values >0.05, Figure 7).

Figure 7 – Energy balance following corticosterone replacement. Consistent with previous experiments, CIT animals had decreased body weight, food intake, and spine length, and increased energy expenditure relative to Air-controls. However, corticosterone replacement during either exposure (CIT-CE) or abstinence (CIT-CA) caused any differentiation between the two CIT groups for any of these variables

Discussion The faecal microbiome results may lead to further avenues of research, and indicate whether or not the gut should be an organ of interest following inhalant abuse, but the lack of results is not central to the research question. Quantification of whole body thermogenesis and physical activity would be relevant to the research aims of this thesis, as they enable a more detailed understanding of the effects of inhalant abuse on the energy balance equation. Due to experimental constraints, the measure of physical activity within the metabolic cages could not be repeated, and this is acknowledged within the limitations and future work section of the thesis. Measurement of whole body thermogenesis via thermographic imaging would be a

125 Rose Crossin (737900) valuable methodology, as it is a non-invasive method that allows for repeated measures in animals. Overall, variability in this method was high, and temperatures were affected by positioning of the animals, who were not sedated. Consideration was given to restraining the animals to get better images, but this was rejected as the stress of restraint may affect body temperature and confound the results. Though the experiments in chapter 4 focussed largely on the adrenal gland, the liver remains a key organ of interest in relation to inhalant abuse, and further research is required (including assessment of liver lipids), which is discussed in the future work section.

Assessment of bone age is not typically conducted using µCT, rather using histology, but given that the tibiae images were already processed, an imaging method was trialled. This was not central to the research question of this thesis, but results may have provided support for the adrenal insufficiency hypothesis. Corticosterone treatment was an important aspect to trial as part of identifying adrenal insufficiency, however, had never been trialled in an inhalant abuse / toluene exposure model. Therefore, a dose was selected that was physiologically active, but was unlikely to result in the development of obesity [156]. Due to experimental constraints, only one dose of corticosterone could be trialled, thus it cannot be determined if the selected dose was too low to overcome the effects of toluene in the CIT model, or whether alternative hypotheses need to be considered. These are discussed in detail in the general discussion. Overall, these findings suggest avenues for future work, but do not fundamentally alter the conclusions of this thesis.

126 Rose Crossin (737900)

Chapter 5 - General Discussion Inhalant abuse is a worldwide public health issue in which adolescents, and particularly those from disadvantaged populations (including indigenous peoples), are disproportionately represented [27, 32-35]. Since the 1960’s it has been known that those who misuse inhalants present as emaciated [24], with later research expanding upon this finding and highlighting that inhalant abuse results in appetite suppression [23, 25, 26], to the extent that these are listed as warning signs of inhalant abuse [44]. This suggests that inhalant abuse has an effect on energy balance, which is of concern given that the peak period of inhalant abuse co-occurs with a high growth period (the adolescent growth spurt), and any disruption to energy balance at this time could have consequences into adulthood. However, despite this knowledge, the effects of adolescent inhalant abuse on energy balance and growth effects have never been fully characterised. Furthermore, despite most inhalant users moving away from inhalants as age increases [27, 33] and thus moving into extended periods of abstinence, the long-term impacts on energy balance and growth remain unknown. Lastly, there is little information regarding the mechanisms underlying the observed weight and energy balance effects of inhalants; a major barrier to developing treatments for these health consequences. These three knowledge gaps formed the major aims of this thesis.

Adolescent inhalant abuse results in growth suppression and energy balance alterations Initially, the growth consequences of adolescent inhalant abuse were explored by undertaking a retrospective analysis of data collected from a cohort of Indigenous Australian males, who chronically sniffed petrol during adolescence. It was found that active inhalant abuse (petrol sniffing in this cohort) resulted in significant impairments to both body weight and height, compared to age and gender-matched controls from the same communities, which enabled the potential confounds of environment and socio-economic status to be controlled for. The findings regarding body weight were consistent with previous clinical and pre-clinical studies [23-25], but this was the first clinical study that could be found at the time of writing that showed height impairments arising from adolescent inhalant abuse. In addition, a link was observed between the duration of petrol sniffing and the degree of height impairment; suggestive of a dose-response relationship. A previous study had found a non-significant effect of inhalant abuse on height in a cohort of adolescent males who chronically sniffed glue [48]; however, the numbers in this study were small and may not have been sufficiently powered to detect changes.

The weight impairments were so severe in this study that adolescent inhalant abuse was significantly associated with FTT, defined as being under the 3rd percentile of weight for age on standard growth charts [157, 158]. FTT is more prevalent in disadvantaged and Indigenous communities generally [157] and a small number of non-petrol sniffing control participants also met FTT criteria, however, this was the first study to specifically link inhalant abuse to FTT. It was also found that FTT could be used a diagnostic measure for identifying individuals who were misusing inhalants; an important finding given that inhalant abuse is notoriously difficult to identify [12], and there are no established diagnostic tests for inhalant abuse. Using FTT as a means of identifying inhalant abuse requires no specialist equipment or training, and thus may be useful for identifying individuals misusing inhalants in remote and disadvantaged

127 Rose Crossin (737900) communities where inhalant abuse is prevalent. Additionally, FTT is associated with significant other health issues including cognitive impairment [159] and delayed skeletal maturation [160]. Height impairment can also lead to adverse psychological outcomes and is associated with issues such as poor self-image and social anxiety [134, 161]. Thus, these growth impairments have the potential to lead to significant other health risks for those who misuse inhalants. The need for early diagnosis and intervention was also highlighted by the finding that inhalant abuse was associated with earlier and increased use of other drugs; by identifying and intervening with individuals early, this trajectory from adolescent inhalant abuse to other substances of abuse may be altered.

Though the observed association with FTT and height impairment were novel findings, there were a number of limitations that should be mentioned. The study was designed and data were collected by others, and the primary aim of the data collection was to consider the social, cognitive, and neurological impacts of inhalant abuse, rather than to assess the growth impacts of adolescent inhalant abuse. By design, because of the predominant population misusing inhalants in the communities of interest, the cohort was only male, all participants were Australian Indigenous, and participants only sniffed petrol rather than using a range of inhalants containing toluene. Thus, they were not representative of the total inhalant abuse population, which is approximately 50% female [27], and who use a range of toluene-based inhalants based on availability [162]. Furthermore, due to the primary research questions underlying the initial project, the data collected from this cohort did not include any measures of body composition or visceral adiposity (e.g. waist circumference), food intake, or effects of inhalant abuse on appetite. Thus, mechanistic interpretation as to the cause of the growth impairments could not be made. Finally, given that at this time it seemed that the observed height impairment was a novel finding in an inhalant abuse clinical cohort (though one study was later identified [163] when conducting the systematic review), it was important to identify whether this finding was consistently observed.

In response to these limitations, and to better understand the growth impacts of inhalant abuse, a systematic review and meta-analysis of human and animal studies was conducted, to investigate the effects of inhalant abuse (in humans) or toluene exposure (in pre-clinical models) on body weight and height. Through the systematic review process only 31 studies on either inhalant abuse or toluene exposure were identified that reported a comparison of treatment-to-control mean for weight and / or height along with a measure of variance. This highlights the paucity of data in this field, which is particularly concerning given the potential health effects arising from changes to growth in adolescence, as outlined earlier. It was found that inhalant abuse / toluene exposure had an overall medium-large effect size on height, confirming findings from the epidemiological study. However, due to the very low number of treatment-to-control comparisons for height, no further analysis on the effects of toluene concentration or study characteristics could be undertaken. This study also showed that inhalant abuse / toluene exposure had an overall medium-large effect size on body weight, again consistent with the epidemiological findings, and that furthermore, there was a concentration-response relationship; with increased concentrations of toluene resulting in more severe impairments to body weight. This was the first meta-analysis on the effects of inhalant abuse on growth measures in the exposed individual, however, there had been a meta-analysis conducted on the effect of pre-natal exposure to toluene on developmental

128 Rose Crossin (737900) birth weight of the offspring. In this study, the authors showed that pre-natal toluene exposure impaired developmental body weight in the offspring, but did not show a linear concentration-response relationship for developmental body weight [46].

The meta-analysis also revealed differences in the effect size on body weight, depending on study characteristics; however, a meta-regression could not be undertaken due to the low number of studies. Body weight impairments differed by species, method of exposure, age at first exposure, and sex. There was similarity in the effect sizes between humans and rats. Data on guinea pigs, dogs and monkeys could not be interpreted due to very low study numbers; however, in studies using mice the effect size on body weight was near zero. It has been previously suggested that toluene exposure may increase body weight and food intake in mice [46], in contrast to findings in both humans [24, 26] and rats [23, 25]. This may limit their utility as a pre-clinical model in which to study the energy balance effects of inhalant abuse, and energy balance outcomes in toluene exposure studies using mice should be interpreted with caution. Similarly, differences were observed in the method of toluene exposure. The majority of pre-clinical studies of inhalant abuse use an inhalation method to administer toluene, and these findings were consistent with humans, though toluene administered via intraperitoneal injection had an effect size near zero and may not be an appropriate administration method for studies investigating the energy balance and growth effects of toluene.

As expected, the age at first exposure differentiated the effects on body weight, with impairments more severe in adolescence when growth is rapidly increasing, compared to adulthood [49], suggesting that the energy balance and growth consequences will be more relevant when inhalant abuse occurs in adolescents, and thus adolescents remain the key population of interest for this research question. Sex-differences in the meta-analysis were observed, with a greater effect size on body weight observed in males. This is in contrast to one of the few human cohorts of inhalant users that had both male and female participants, which showed that the effect on body weight is more severe in adolescent females [164]. This issue requires further investigation, because females were highly under-represented in the meta-analysis. However, body weight changes during adolescence are particularly relevant to females, for whom a disruption to body weight at this time may impact pubertal processes, menarche, and affect future fertility [165, 166].

Interpretation of findings from the meta-analysis was limited by inconsistencies in the available data across sub-groups. It is therefore possible that the observed differences in the meta-analysis between clinical studies and pre-clinical models may be an artefact of the data availability. For example, differences were found in the concentrations of toluene that different sub-groups within each study characteristic were exposed to. In the highest inhaled concentration sub-group of >5,000 parts per million (ppm), there were only males and no females, only adolescents and no adults, and only rats and no other animal species. Given that higher effect sizes were observed in males, adolescents, and rats, it is possible that this is simply reflecting the response to the high concentration of toluene, rather than sex, age, and species moderating the relationship between toluene exposure and growth changes. The meta-analysis was limited by the low number of studies, and thus it may be valuable to repeat this study in the future as the body of literature grows. The main limitation of this study is that, by its nature, a meta-analysis cannot identify a mechanism by which inhalant abuse affects

129 Rose Crossin (737900) growth, and while this study showed that height and weight impairments are significant following adolescent inhalant abuse, the mechanism underlying these changes remained unknown.

From the epidemiological study and meta-analysis there was sufficient evidence that adolescent inhalant abuse significantly impairs both body weight and height. However, there were still significant knowledge gaps. For example, body weight is a function of energy stored in the body, which is determined by the energy balance equation: energy balance = energy intake - energy expenditure (see Figure 2). Though the findings of suppressed body weight and food intake [23-26] after adolescent inhalant abuse suggested that there was an energy balance effect, the variables within the energy balance equation had not been quantified. Using an established rodent model that recapitulates human inhalant abuse (called the chronic intermittent toluene, or CIT model), it was found that toluene exposure to adolescent male rats significantly impaired both body weight and spine length (analogous to human height), consistent with both the epidemiological study and meta-analysis. The finding that body weight was significantly impaired following adolescent toluene exposure was consistent with previous pre-clinical findings [23, 25]. However, growth (rump width and spine length) was also measured, which was also suppressed following toluene exposure, suggesting that toluene has an effect on linear growth as well as body weight. The energy balance effects were characterised by assessing the variables within the energy balance equation, which had not been previously undertaken. It was found that adolescent inhalant abuse induced a significant negative energy balance phenotype, with concomitant decreases in energy intake and increases in energy expenditure at the end of the exposure period in the CIT group. Energy expenditure had not been assessed in previous studies of inhalant abuse, and these findings highlight the importance of quantifying both sides of the energy balance equation when growth changes are observed. The finding of suppressed food intake during the exposure period was consistent with previous studies that had found that exposure to toluene during adolescence significantly and acutely decreased food intake [23, 25], however, these findings were added to by conducting faecal bomb calorimetry that revealed that toluene exposure did not affect absorption of food in the gut, at the end of exposure. This implies that the changes to energy intake within the energy balance equation are due to food intake, rather than changes to absorption of food in the gut.

The growth impairments arising from adolescent inhalant abuse persist into sustained abstinence In order to understand the long-term consequences of adolescent inhalant abuse on growth, it is important to determine whether or not the observed changes resolve or persist in abstinence. To address this, data were analysed from a sub-set of the same cohort of Australian Indigenous males, who had been abstinent from inhalant abuse for two years, following interventions that eradicated petrol sniffing from their communities [151, 167]. It was found that the height impairments that had arisen from inhalant abuse had not resolved in abstinence, and although study participants had continued to grow, no catch-up growth was evident. This result is consistent with knowledge about the adolescent growth spurt, whereas disruptions to growth in childhood can be recovered if the factor limiting growth is removed, but the potential for catch-up growth in adolescence ranges from partial to nil [54]. These

130 Rose Crossin (737900) findings were recapitulated in the rodent model, in which animals exposed to toluene for four weeks (CIT) throughout adolescence remained significantly shorter than those not exposed to toluene, at the end of the four week abstinence period. The potential for permanent height impairment is a powerful public health message for young teens, particularly in Australian Indigenous communities, where size and strength are highly valued personal attributes [168]. This finding gained media attention when it was released in 2017 and was used by Indigenous health services in the Northern Territory as a deterrent message in populations where there was a recent resurgence in inhalant abuse.

In contrast to the findings for height, in the human cohort, it was found that body weight of the (ex) petrol sniffing group had normalised to the non-petrol sniffing controls following two years of abstinence. This was also inconsistent with the findings from the rodent model, in which body weight in the CIT group remained significantly impaired in sustained abstinence (at an age equivalent to the human cohort), despite normalisation of food intake, consistent with previous findings in male adolescent rats [23]. It is well understood that weight impairments are homeostatically corrected for [58], and thus it would be expected that body weight would normalise if the factor suppressing body weight was removed. However, there are potentially some confounds within the epidemiology data, particularly due to other substance use. Though petrol sniffing was eradicated from these communities, other drug use, particularly the use of cannabis and alcohol increased [152]. Both of these drugs have been associated with weight gain in humans [169, 170], and thus could be contributing to weight gain in abstinence, though this could not be tested statistically in this study due to insufficient numbers of ex- petrol sniffing individuals who had no additional other substance use. It is also possible that differences in homeostatic recovery between humans and rats may account for these different results, and this will require re-testing in a human cohort who is not only abstinence from inhalants, but also other substances of abuse. It also cannot be determined from this study whether the weight gain is a healthy regain of energy balance homeostasis following a period of weight impairment, or whether the rapid weight gain (an average of 9 kg in 2 years) may be a precursor to visceral adiposity, insulin resistance, and future metabolic disorders [136]. Therefore, the implications for long-term health arising from this weight gain remain unknown.

Growth changes arising from adolescent inhalant abuse are not attributable to under-nutrition but may be caused by adrenal insufficiency Potential mechanisms underlying the observed growth consequences of adolescent inhalant abuse were explored in the rodent CIT model. Given it is known that inhalant abuse suppresses food intake [23, 25, 26], it was a reasonable starting point to hypothesise that the growth changes are a response to under-nutrition, causing a negative energy balance. This was an important idea to test, because from a clinical perspective, if changes to growth are driven by under-nutrition, then nutritional intervention may be an effective treatment. However, evidence that under-nutrition is the sole mediator of inhalant-induced changes in growth was limited. Previous studies had shown that exposure to toluene during adolescence altered the relationship between food intake and body weight, and caused metabolic dysfunction including non-diabetic fasting hypoglycaemia and decreased adiposity, suggestive of a broader effect on energy balance [23]. Given that reduced food intake can induce negative energy

131 Rose Crossin (737900) balance and thus cause a loss of body weight, previous experiments had not determined if the effects of toluene on body weight were a direct effect, or an indirect effect arising from toluene-induced under-nutrition. Therefore, the under-nutrition hypothesis was tested via the inclusion of a pair-fed (to the CIT) group who were not exposed to toluene. This allowed separation of the direct effects of toluene exposure (the CIT group) from the secondary effects arising from toluene-induced under-nutrition (the pair-fed group).

Significant differences were found between the pair-fed and CIT groups, which strongly suggest that under-nutrition is not the primary driver of growth changes following toluene exposure. There were three main differences; normalisation of body weight / growth, skeletal effects, and glycaemic effects. Consistent with the CIT group, the pair-fed group showed suppressed weight and growth during the exposure period, when food intake was decreased (reduced by up to 26% at the end of exposure). However, the CIT group’s food intake normalised in abstinence consistent with previous studies [23], and when this occurred, the pair-fed group’s weight and growth rapidly re-normalised to Air controls. This is consistent with homeostatic correction, following a period of under-nutrition [58], but was not observed in the CIT group, where both body weight and growth remained significantly impaired. This suggested that the CIT group may have an impairment to energy balance signalling pathways. Secondly, the pair-fed group showed trabecular bone loss at the end of the exposure period that was consistent with malnutrition [171], but this was not observed in the CIT group.

The effects of malnutrition on bone are primarily an indirect endocrine effect [62-64], including a stress response arising from low food intake [172], and so the absence of this finding in the CIT group suggested that a broader disruption to either energy balance signalling and / or the stress response may be occurring. Finally, the CIT group showed non-diabetic fasting hypoglycaemia at the end of the exposure period, consistent with previous studies [23], but this did not occur in the pair-fed group, who were able to maintain blood glucose levels despite significant under-nutrition. The CIT group however had equivalent liver glycogen stores to both the Air control and pair-fed groups, suggesting that disruption to glycogenolysis was occurring. This finding suggested a broader disruption to glycaemic control or the ability to sense falling blood glucose levels, or impairment in the stress response that should be quickly activated in response to low blood glucose [65, 66]. Common themes emerging from these findings are that toluene exposure may cause impairments to energy balance signalling, and impairments to the stress response that should occur in response to low food intake and blood glucose levels. The implication of these being that homeostatic correction following a negative energy balance may not occur, and thus, may persist even after toluene exposure has ceased. One limitation of these experiments is that a pair-fed group was unable to be used when investigating energy expenditure due to experimental constraints, so interpretation of the finding that toluene exposure caused a significant increase in energy expenditure, in the context of under-nutrition, is limited. However, it is well-understood that energy expenditure is typically decreased during periods of under-nutrition, as a means of conserving body stores [173], so this would again suggest that under-nutrition is not the primary driver of these changes. Collectively, these findings suggested that alternative causal hypotheses for the observed growth impairments needed to be considered.

132 Rose Crossin (737900)

Disruption to the HPA axis, and particularly the adrenal gland was a plausible alternative hypothesis. Findings from the previous experiments had suggested that stress responses may be impaired, and multiple previous studies had identified adrenal hypertrophy as an outcome of toluene exposure [23, 126, 127]; therefore, the adrenal gland was focussed on specifically, rather than the HPA axis. Adrenal hypertrophy can, however, be an outcome of either chronic stress or adrenal insufficiency [129], and previous studies had attributed this finding to chronic stress [127], despite the endocrine signature of high ACTH with either low or unchanged corticosterone being more consistent with primary adrenal insufficiency [129, 130]. Adrenal structure and HPA axis signalling and responsivity were tested at the end of both exposure and abstinence, using a diagnostic flowchart approach (Figure 4 of the general introduction), and findings were consistent with adrenal insufficiency at the end of both the exposure and abstinence periods in the CIT group. However, a treatment for adrenal insufficiency was trialled with corticosterone in the drinking water (25 mg/L) during both the exposure period (to try to prevent toluene-induced energy balance changes) and abstinence period (to try to ameliorate toluene-induced energy balance changes), using a crossover design. This treatment was ineffective during both the exposure and abstinence periods. This does not however provide sufficient evidence to reject the adrenal insufficiency hypothesis. One plausible explanation is that, despite this dose having been previously shown to partially correct changes to metabolic neuropeptides caused by adrenalectomy [155], the dose was too low to counteract the effects of toluene in this model. An alternative explanation is that glucocorticoid resistance may be concurrent with adrenal insufficiency [174]. Support for the glucocorticoid resistance hypothesis was provided via a stress response test, which showed that in the toluene exposed animals it took double the amount of corticosterone release to achieve an equivalent shift in blood sugar levels. However, further testing of this hypothesis was outside the scope of this research due to timing constraints.

The mechanistic experiments advanced understanding of the causes of the energy balance and growth consequences of adolescent inhalant abuse. Through the use of a pair-fed group, a potential hypothesis for the observed growth changes (i.e. under-nutrition) was able to be rejected, and experiments highlighted that toluene was having a direct effect on growth, with evidence suggesting impairments to energy balance signalling and/or stress responsivity. Evidence was provided for an alternative hypothesis (adrenal insufficiency) for the observed growth changes; importantly, findings were consistent with adrenal insufficiency at the end of the exposure period and following sustained abstinence. The utility of these data is that they highlight potential long-term health risks for those with a history of adolescent inhalant abuse; both growth impairments and adrenal insufficiency. In addition to the health risks of adrenal insufficiency itself, including the potential for a life-threatening adrenal crisis [130], this finding may provide a novel target for intervention, to address the known link between inhalant abuse and suicidality [38, 39], as blunted HPA axis responsivity, as observed in these experiments, has been associated with increased suicidal behaviours [175, 176]. There are additional studies and replication needed in a pre-clinical model before these findings can be confirmed, which will be discussed in more detail below. Additionally, as the study was conducted in a pre- clinical model it will need to be explored in a clinical setting before the implications of this research can be fully understood.

133 Rose Crossin (737900)

Translational applications of this research Adolescent inhalant abuse significantly impairs body weight, to the extent that it is associated with FTT. Furthermore, FTT can be used as a diagnostic marker for inhalant abuse, which is particularly useful in vulnerable and remote populations where inhalant abuse is prevalent. Thus, it would be beneficial to amend the differential diagnosis for FTT, in order to include inhalant abuse in high risk populations and age groups such as adolescents. Adolescent inhalant abuse also significantly impairs height and this persists into sustained abstinence. It would therefore be beneficial to include height impairments as a warning sign of inhalant abuse. The benefits of this are two-fold. Firstly, it may improve identification of inhalant abuse, if a parent, caregiver, or primary health care professional notices that height trajectory in an individual has changed. Secondly, it may be a powerful deterrent message to adolescents, for whom persistent height impairments may be an undesirable outcome. While it was found that adolescent inhalant abuse results in a negative energy balance phenotype and adrenal dysfunction consistent with adrenal insufficiency, this research requires clinical confirmation before it can be fully utilised in order to improve treatment of those with a history of inhalant abuse. However, given that pair-feeding studies showed that the growth changes are independent of food intake changes, interventions that seek to increase food intake are unlikely to be effective as treatments for resolving the observed growth impairments. These findings suggest that potential treatments targeting adrenal function may be beneficial to prevent long-term health impacts in those with a history of adolescent inhalant abuse.

The findings from the systematic review and meta-analysis may also have utility for the inhalant abuse field, as the overall lack of data and also the imbalance of data was highlighted. As discussed, the lack of data on adolescent females is particularly concerning, because of the potential for reduced body weight and adiposity to impact both future reproductive capacity [165, 166] and fetal health by increasing the risk of pre-term birth and low birthweight [177]. Also of concern is the lack of data on the effects of exposure to high concentrations of toluene, in species other than rats and humans that may be used in pre-clinical studies. It is also important for the inhalant abuse field that length / height be measured more often as an outcome, along with body weight. An observation of normal height with emaciation is physiologically different from an observation of both stunting and concurrent emaciation, but these cannot be differentiated unless both body weight and height are measured.

Future work This research has advanced our understanding of the growth and energy balance consequences of adolescent inhalant abuse, however, future work remains, some of which will be best answered in pre-clinical models. Though adolescent inhalant abuse induces a negative energy balance phenotype, it is still unknown why toluene suppresses appetite and food intake from the perspective of neural circuitry and energy balance signalling that regulates food intake. It would be beneficial for the neural circuitry and peripheral signalling that regulates food intake and energy balance status to be investigated in detail, particularly in the hypothalamus, following adolescent inhalant abuse, which will provide greater knowledge into the effects of inhalants and toluene. The adrenal gland was investigated as a component of the HPA axis, however, detailed studies into the hypothalamus were beyond the scope of this thesis, but would provide valuable further knowledge. It was found that inhalant abuse

134 Rose Crossin (737900) increases energy expenditure, however, this could not be differentiated by the variables that make up energy expenditure (i.e. physical activity, basal metabolic rate, and thermogenesis). It would be beneficial for energy expenditure in this model to be explored in more detail, including with a pair-fed group, and to also determine the persistence of these effects into sustained abstinence. A method for measuring whole body thermogenesis was trialled, using thermographic imaging, however, these data were highly inconsistent within individual subjects (even in images taken seconds apart), and thus potential changes to thermogenesis may have been undetected using this method. Unless modifications to this protocol can be made, understanding of thermogenesis will be best obtained by UCP1 analysis in brown adipose tissue and inguinal white adipose tissue (where “beiging” is prevalent) [178]. The metabolic cage protocol used should have provided data on physical activity to pair with metabolic rate, however, due to an equipment failure this variable was not obtained. Repeating this experiment should yield the required data, and enable quantification of physical activity, as a variable contributing to overall energy expenditure.

In relation to the HPA axis and adrenal insufficiency findings, a number of questions remain. Adrenal structure and function was measured at two time points (i.e. the end of exposure and the end of abstinence). However, the functionality of the stress response system in the intervening periods cannot be determined. It is possible that there was a linear decline in HPA axis function beginning from the start of the exposure period. It is also possible that there was a dramatic increase in HPA axis activity during the exposure period, followed by a decline in function as this system became dysfunctional or de-sensitised [179]. However, the experimental design did not allow for differentiation between these scenarios. To clarify this issue, it would be beneficial to gather detailed temporal data on HPA axis activity as well as related anxiety or stress-related behaviours, throughout the course of the exposure and abstinence periods. As these data also suggested that glucocorticoid resistance may be occurring in addition to adrenal insufficiency, and may explain why corticosterone treatment (25 mg/L in drinking water) was ineffective, during both the exposure and abstinence periods, thus glucocorticoid signalling and responsivity should be measured in key metabolic organs such as the pancreas and liver. It would also be beneficial to trial higher doses of corticosterone, as it is possible that the trialled dose was insufficient to overcome the effects of toluene. Corticosterone treatment had never before been used in this model, so a dose was selected that would be physiologically active [180, 181], but not so high that it promoted excessive body weight gain or adiposity [156], but due to experimental constraints, only a single dose could be trialled. More broadly, the effects of inhalant abuse on the HPA axis are highly equivocal [119-121, 123, 126, 127], but this may be in part due to the issues highlighted in the meta-analysis i.e. responses may be altered in response to toluene concentration, exposure patterns, and study characteristics. This topic would benefit from a systematic characterisation approach, assessing HPA axis signalling, functionality, and associated behaviours, within various study characteristics such as sex, age, and toluene concentration and exposure patterns. The inclusion of a pair-fed group may yield relevant data on the role of under-nutrition on HPA axis function. Finally, all of this work, including the energy balance and HPA axis / adrenal experiments should be repeated in adolescent females. Adolescent males were used in the pre-clinical experiments to provide continuity from the epidemiology studies, but it should not be assumed that these findings can be translated to females.

135 Rose Crossin (737900)

In addition to this suggested pre-clinical research, some of these findings would be best utilised by translation and validation in clinical studies. It would be valuable to validate the energy balance findings, including both energy intake and expenditure, in a human cohort; a study that has started but as yet has insufficient data to draw conclusions. This study will include assessment of body composition, and metabolic variables such as insulin and leptin levels, as well as assessment of fasting blood glucose levels. Importantly, this cohort will seek to recruit female inhalant users as well as males, to more accurately represent the inhalant abuse population. Preliminary results (Appendix 2) suggest that this study will be consistent with findings from Chapters 2-4, with participants to date having growth impairments and changes to food intake and appetite. It would also be beneficial to clinically assess the presence of adrenal insufficiency in an inhalant abuse cohort, by conducting testing of basal cortisol and ACTH levels, as well as dynamic testing such as an insulin tolerance test. This would take a cohort approach to the issue by comparing adrenal insufficiency occurrence following adolescent inhalant abuse to a population who did not misuse inhalants. An alternative or complementary approach could be to take a case-control approach and use a population of those with adrenal insufficiency and investigate the prevalence of historical inhalant abuse. Human cohorts of inhalant users are very rare, but future cohorts should attempt to include equal numbers of males and females, and to maintain the cohort longitudinally into abstinence. Both the energy balance and adrenal impacts of inhalant abuse need to be assessed into sustained abstinence, in order to understand the long-term health impacts of this behaviour. For this, longitudinal cohorts of inhalant users will be vital.

Summary and conclusions Inhalant abuse is a relatively under-studied form of substance abuse, which is prevalent in adolescents, particularly those in vulnerable or disadvantaged populations; these patterns are consistent worldwide. Such populations already have poorer health outcomes on average [182], and thus it is concerning that the energy balance effects of inhalant abuse had remained poorly understood since the 1960’s, despite this being a highly visible change with the potential for long-term health impacts even if inhalant abuse ceases.

Collectively, these studies found that adolescent inhalant abuse causes significant impairments to both body weight and height, which persist into sustained abstinence. Furthermore, inhalant abuse induces a negative energy balance phenotype, with energy intake decreased simultaneously with increases to energy expenditure. However, under-nutrition resulting from reduced food intake did not causally underpin the observed growth changes. Instead, it was found that adrenal insufficiency can arise from adolescent inhalant abuse, and that adrenal dysfunction may persist into sustained abstinence, which may mechanistically explain the observed growth impairments. The adrenal insufficiency hypothesis requires testing in a clinical setting, but if confirmed, presents a significant health risk to those with a history of inhalant abuse, as the disorder can be life-threatening when undiagnosed and untreated.

This research has contributed to the inhalant abuse field, by systematically characterising the well-known effects of inhalant abuse (i.e. weight loss and appetite suppression) within the context of the energy balance equation, and by the discovery of previously unknown consequences of adolescent inhalant abuse. This has provided targets for future clinical research, which may improve the health outcomes of highly vulnerable individuals.

136 Rose Crossin (737900)

Furthermore, knowledge specific to pre-clinical inhalant abuse field has been provided, highlighting issues for consideration when developing and using pre-clinical models, which may improve knowledge translation in this field. Collectively, this research has utilised multiple methods both clinical and pre-clinical, in order to better understand the health consequences of adolescent inhalant abuse, and to provide translational outputs that will directly improve the detection and treatment of inhalant abuse in adolescents. In conclusion, adolescent inhalant abuse affects energy balance, and has long-term effects on growth, which may continue to affect the long-term health of individuals even if inhalant abuse ceases.

137 Rose Crossin (737900)

Bibliography 1. National Institute on Drug Abuse. What is drug addiction? 2014 July 2014 [cited 2015 14/7/2015]; Available from: http://www.drugabuse.gov/publications/drugs-brains- behavior-science-addiction/drug-abuse-addiction. 2. Alcohol and other Drugs Council of Australia, Policy Position - Inhalants. 2010, Alcohol and other Drugs Council of Australia: Canberra. 3. Victorian Department of Human Services, National Directions on Inhalant Abuse. 2005, Victorian Department of Human Services: Melbourne. 4. Duncan, J.R. and A.J. Lawrence, Conventional concepts and new perspectives for understanding the addictive properties of inhalants. Journal of Pharmacological Sciences, 2013. 122(4): p. 237-243. 5. Funada, M., et al., Evaluation of rewarding effect of toluene by the conditioned place preference procedure in mice. Brain Research Protocols, 2002. 10(1): p. 47-54. 6. Villatoro, J.A., et al., Volatile substance misuse in Mexico: correlates and trends. Substance Use & Misuse, 2011. 46(sup1): p. 40-45. 7. Bowen, S.E., et al., Decreased sensitivity in adolescent vs. adult rats to the locomotor activating effects of toluene. Neurotoxicology and Teratology, 2007. 29(6): p. 599-606. 8. Dick, A.L., et al., Specific impairments in instrumental learning following chronic intermittent toluene inhalation in adolescent rats. Psychopharmacology, 2014. 231(8): p. 1531-1542. 9. Pryor, G.T., A toluene-induced motor syndrome in rats resembling that seen in some human solvent abusers. Neurotoxicology and Teratology, 1991. 13(4): p. 387-400. 10. Sharp, C. and N. Rosenberg, Volatile substances. 2nd ed. Substance Abuse: A Comprehensive Textbook. 1992, Baltimore: Williams & Wilkins. P. 222-235. 11. Bowen, S.E., et al., The last decade of solvent research in animal models of abuse: mechanistic and behavioral studies. Neurotoxicology and Teratology, 2006. 28(6): p. 636-647. 12. Kurtzman, T.L., K.N. Otsuka, and R.A. Wahl, Inhalant abuse by adolescents. Journal of Adolescent Health, 2001. 28(3): p. 170-180. 13. Riegel, A., S. Ali, and E. French, Toluene-induced locomotor activity is blocked by 6- hydroxydopamine lesions of the nucleus accumbens and the mGluR2/3 agonist LY379268. Neuropsychopharmacology, 2003. 28(8): p. 1440-1447. 14. Evans, E.B. and R.L. Balster, CNS depressant effects of volatile organic solvents. Neuroscience & Biobehavioral Reviews, 1991. 15(2): p. 233-241. 15. Shepherd, R., Mechanism of sudden death associated with volatile substance abuse. Human & Experimental Toxicology, 1989. 8(4): p. 287-291. 16. Sato, A. and T. Nakajima, Pharmacokinetics of organic solvent vapors in relation to their toxicity. Scandinavian Journal of Work, Environment & Health, 1987: p. 81-93. 17. Sato, A., et al., Pharmacokinetics of benzene and toluene. Internationales Archiv für Arbeitsmedizin, 1974. 33(3): p. 169-182. 18. Lubman, D., M. Yücel, and A. Lawrence, Inhalant abuse among adolescents: neurobiological considerations. British Journal of Pharmacology, 2008. 154(2): p. 316- 326. 19. Hormes, J.T., C.M. Filley, and N.L. Rosenberg, Neurologic sequelae of chronic solvent vapor abuse. Neurology, 1986. 36(5): p. 698-698. 20. Knox, J.W. and J.R. Nelson, Permanent encephalopathy from toluene inhalation. New England Journal of Medicine, 1966. 275(26): p. 1494-1496. 21. Maruff, P., et al., Neurological and cognitive abnormalities associated with chronic petrol sniffing. Brain, 1998. 121(10): p. 1903-1917.

138 Rose Crossin (737900)

22. Pyykkö, K., H. Tähti, and H. Vapaatalo, Toluene concentrations in various tissues of rats after inhalation and oral administration. Archives of Toxicology, 1977. 38(3): p. 169- 176. 23. Dick, A.S., Annabeth; Qama, Ashleigh; Andrews, Zane; Lawrence, Andrew John; Duncan, Jhodie Rubina, Chronic intermittent toluene inhalation in adolescent rats results in metabolic dysfunction with altered glucose homeostasis. British Journal of Pharmacology, 2015. 172(21): p. 5174-5187. 24. Glaser, H.H. and O.N. Massengale, Glue-sniffing in children: Deliberate inhalation of vaporized plastic cements. Journal of the American Medical Association, 1962. 181(4): p. 300-303. 25. Morón, L., et al., Toluene alters appetite, NPY, and galanin immunostaining in the rat hypothalamus. Neurotoxicology and Teratology, 2004. 26(2): p. 195-200. 26. Pisetsky, E.M., et al., Disordered eating and substance use in high‐school students: Results from the Youth Risk Behavior Surveillance System. International Journal of Eating Disorders, 2008. 41(5): p. 464-470. 27. White, V. and T. Williams, Australian secondary school students’ use of tobacco, alcohol, and over-the-counter and illicit substances in 2014. 2016, Cancer Council Victoria for Australian Government Department of Health: Melbourne, Australia. 28. Andersen, I., et al., Human response to controlled levels of toluene in six-hour exposures. Scandinavian Journal of Work, Environment & Health, 1983: p. 405-418. 29. Dick, R.B., et al., Effects of acute exposure of toluene and methyl ethyl ketone on psychomotor performance. International Archives of Occupational and Environmental Health, 1984. 54(2): p. 91-109. 30. Green, M., Bright Futures: Guidelines for Health Supervision of Infants, Children, and Adolescents. 1994: ERIC. 31. Australian Institute of Health and Welfare, National Drug Strategy Household Survey 2016. 2016, Australian Institute of Health and Welfare: Canberra. 32. Dell, C.A., S.W. Gust, and S. MacLean, Global issues in volatile substance misuse. Substance Use & Misuse, 2011. 46(sup1): p. 1-7. 33. Johnston, L.D., et al., Monitoring the Future - Key Findings on Adolescent Drug Use 2017 overview. 2017, The University of Michigan: Ann Arbor. 34. Tapia‐Conyer, R., et al., Risk factors for inhalant abuse in juvenile offenders: the case of Mexico. Addiction, 1995. 90(1): p. 43-49. 35. Australian Institute of Health and Welfare, Substance abuse among Aboriginal and Torres Strait Islander people. 2011, Australian Institute of Health and Welfare: Canberra. 36. Cairney, S., et al., The neurobehavioural consequences of petrol (gasoline) sniffing. Neuroscience & Biobehavioral Reviews, 2002. 26(1): p. 81-89. 37. Wu, L.-T., M.O. Howard, and D.J. Pilowsky, Substance use disorders among inhalant users: Results from the National Epidemiologic Survey on Alcohol and Related Conditions. Addictive Behaviors, 2008. 33(7): p. 968-973. 38. Sakai, J.T., et al., Inhalant use, abuse, and dependence among adolescent patients: commonly comorbid problems. Journal of the American Academy of Child & Adolescent Psychiatry, 2004. 43(9): p. 1080-1088. 39. Howard, M.O., et al., Suicide ideation and attempts among inhalant users: results from the national epidemiologic survey on alcohol and related conditions. Suicide and Life- Threatening Behavior, 2010. 40(3): p. 276-286. 40. Ryu, Y.H., et al., Cerebral perfusion impairment in a patient with toluene abuse. Journal of Nuclear Medicine: Official Publication, Society of Nuclear Medicine, 1998. 39(4): p. 632-633.

139 Rose Crossin (737900)

41. Brady, M., Heavy metal: the social meaning of petrol sniffing in Australia. 1992, Canberra: Aboriginal Studies Press. 42. Jarosz, P.A., et al., Effects of abuse pattern of gestational toluene exposure on metabolism, feeding and body composition. Physiology & Behavior, 2008. 93(4): p. 984-993. 43. Duncan, J.R., et al., Adolescent toluene inhalation in rats affects white matter maturation with the potential for recovery following abstinence. PLoS ONE, 2012. 7(9): p. e44790. 44. Alcohol and Drug Foundation. Drug facts: inhalants. 2015 19/05/2018 [cited 2018 20/05/2018]; Available from: http://www.druginfo.adf.org.au/drug-facts/inhalants. 45. Roberts, L., A. Bevans, and C. Schreiner, Developmental and reproductive toxicity evaluation of toluene vapor in the rat: I. Reproductive toxicity. Reproductive Toxicology, 2003. 17(6): p. 649-658. 46. Callan, S., et al., Changes in developmental body weight as a function of toluene exposure: A meta-analysis of animal studies. Human & Experimental Toxicology, 2016. 35(4): p. 341-352. 47. Pearson, M.A., et al., Toluene embryopathy: delineation of the phenotype and comparison with fetal alcohol syndrome. Pediatrics, 1994. 93(2): p. 211-215. 48. Dündaröz, M.R., S. Sarici, and T. Turkbay, Evaluation of bone mineral density in chronic glue sniffers. The Turkish Journal of Pediatrics, 2002. 44(4): p. 326-329. 49. Tanner, J. and R. Whitehouse, Clinical longitudinal standards for height, weight, height velocity, weight velocity, and stages of puberty. Archives of Disease in Childhood, 1976. 51(3): p. 170-179. 50. Bonjour, J.-P., et al., Critical Years and Stages of Puberty for Spinal and Femoral Bone Mass Accumulation during Adolescence*. The Journal of Clinical Endocrinology & Metabolism, 1991. 73(3): p. 555-563. 51. Gordon, C., Measurement of bone density in children. Current Opinion in Endocrine and Metabolic Research, 2005. 12: p. 444-51. 52. Dreizen, S., C.N. Spirakis, and R.E. Stone, A comparison of skeletal growth and maturation in undernourished and well-nourished girls before and after menarche. The Journal of Pediatrics, 1967. 70(2): p. 256-263. 53. Golub, M.S., et al., Effects of exogenous estrogenic agents on pubertal growth and reproductive system maturation in female rhesus monkeys. Toxicological Sciences, 2003. 74(1): p. 103-113. 54. Martorell, R., L.K. Khan, and D.G. Schroeder, Reversibility of stunting: epidemiological findings in children from developing countries. European Journal of Clinical Nutrition, 1994. 48: p. S45-57. 55. Reed, R.B. and H.C. Stuart, Patterns of growth in height and weight from birth to eighteen years of age. Pediatrics, 1959. 24(5): p. 904-921. 56. Woods, S.C., et al., Signals that regulate food intake and energy homeostasis. Science, 1998. 280(5368): p. 1378-1383. 57. Keesey, R.E. and T.L. Powley, Body energy homeostasis. Appetite, 2008. 51(3): p. 442- 445. 58. Sumithran, P. and J. Proietto, The defence of body weight: a physiological basis for weight regain after weight loss. Clinical Science, 2013. 124(4): p. 231-241. 59. Keesey, R.E., Physiological regulation of body weight and the issue of obesity. The Medical Clinics of North America, 1989. 73(1): p. 15-27. 60. Rogol, A.D., P.A. Clark, and J.N. Roemmich, Growth and pubertal development in children and adolescents: effects of diet and physical activity. The American Journal of Clinical Nutrition, 2000. 72(2): p. 521S-528S.

140 Rose Crossin (737900)

61. Rogol, A.D., J.N. Roemmich, and P.A. Clark, Growth at puberty. Journal of Adolescent Health, 2002. 31(6): p. 192-200. 62. Newman, M.M. and K.A. Halmi, Relationship of bone density to estradiol and cortisol in anorexia nervosa and bulimia. Psychiatry Research, 1989. 29(1): p. 105-112. 63. Munoz, M. and J. Argente, Anorexia nervosa in female adolescents: endocrine and bone mineral density disturbances. European Journal of Endocrinology, 2002. 147(3): p. 275-286. 64. Miller, K.K., Endocrine effects of anorexia nervosa. Endocrinology and Metabolism Clinics of North America, 2013. 42(3): p. 515. 65. Verberne, T., A. Sabetghadam, and W. Korim, Neural pathways that control the glucose counterregulatory response. Frontiers in Neuroscience, 2014. 8: p. 38. 66. Sprague, J.E. and A.M. Arbeláez, Glucose counterregulatory responses to hypoglycemia. Pediatric Endocrinology Reviews, 2011. 9(1): p. 463-475. 67. Woods, S.C., et al., Chronic intracerebroventricular infusion of insulin reduces food intake and body weight of baboons. Nature, 1979. 282: p. 503-505. 68. Pittner, R.A., et al., Molecular physiology of amylin. Journal of Cellular Biochemistry, 1994. 55(S1994A): p. 19-28. 69. Bearn, A., B.H. Billing, and S. Sherlock, The response of the liver to insulin in normal subjects and in diabetes mellitus; hepatic vein catheterisation studies. Clinical Science, 1952. 11(2): p. 152. 70. Rabkin, R., M. Ryan, and W. Duckworth, The renal metabolism of insulin. Diabetologia, 1984. 27(3): p. 351-357. 71. Kojima, M., et al., Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature, 1999. 402(6762): p. 656-660. 72. Woods, S.C., Gastrointestinal satiety signals I. An overview of gastrointestinal signals that influence food intake. American Journal of Physiology-Gastrointestinal and Liver Physiology, 2004. 286(1): p. G7-G13. 73. Batterham, R.L. and S.R. Bloom, The gut hormone peptide YY regulates appetite. Annals of the New York Academy of Sciences, 2003. 994(1): p. 162-168. 74. Zhang, Y., et al., Positional cloning of the mouse obese gene and its human homologue. Nature, 1994. 372(6505): p. 425-432. 75. Buettner, C., et al., Leptin controls adipose tissue lipogenesis via central, STAT3– independent mechanisms. Nature Medicine, 2008. 14(6): p. 667-675. 76. Schwartz, M.W., et al., Central nervous system control of food intake. Nature, 2000. 404(6778): p. 661-671. 77. Morley, J., E. Hernandez, and J. Flood, Neuropeptide Y increases food intake in mice. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology, 1987. 253(3): p. R516-R522. 78. Lee, N.K., et al., Endocrine regulation of energy metabolism by the skeleton. Cell, 2007. 130(3): p. 456-469. 79. Kersten, S., Mechanisms of nutritional and hormonal regulation of lipogenesis. EMBO Reports, 2001. 2(4): p. 282-286. 80. Kennedy, G.C., The role of depot fat in the hypothalamic control of food intake in the rat. Proceedings of the Royal Society of London B: Biological Sciences, 1953. 140(901): p. 578-592. 81. Marieb, E.N. and K. Hoehn, Human anatomy & physiology. 2007, New York: Pearson Education. 82. US Environmental Protection Agency, Toxicological review of toluene. 2005, US Environmental Protection Agency: Washington DC. 83. Gerasimov, M., et al., Study of brain uptake and biodistribution of [11 C] toluene in non- human primates and mice. Life Sciences, 2002. 70(23): p. 2811-2828.

141 Rose Crossin (737900)

84. Mathew, R.J. and W.H. Wilson, Substance abuse and cerebral blood flow. The American Journal of Psychiatry, 1991. 148(3): p. 292. 85. Ameno, K., et al., Regional brain distribution of toluene in rats and in a human autopsy. Archives of Toxicology, 1992. 66(2): p. 153-156. 86. Luiten, P., G. Ter Horst, and A. Steffens, The hypothalamus, intrinsic connections and outflow pathways to the endocrine system in relation to the control of feeding and metabolism. Progress in Neurobiology, 1987. 28(1): p. 1-54. 87. Jackson, C.E., et al., Hunger and the perception of the scent of petrol: A potential neurobiological basis for increased risk of petrol inhalation abuse. Addiction Research & Theory, 2009. 17(5): p. 518-524. 88. Gigengack, R., “My body breaks. I take solution.” Inhalant use in Delhi as pleasure seeking at a cost. International Journal of Drug Policy, 2014. 25(4): p. 810-818. 89. Bode, C. and J.C. Bode, Effect of alcohol consumption on the gut. Best Practice & Research Clinical Gastroenterology, 2003. 17(4): p. 575-592. 90. Anderson, C.E. and G.A. Loomis, Recognition and prevention of inhalant abuse. American Family Physician, 2003. 68(5): p. 869-874. 91. Batis, J.C., J.H. Hannigan, and S.E. Bowen, Differential effects of inhaled toluene on locomotor activity in adolescent and adult rats. Pharmacology Biochemistry and Behavior, 2010. 96(4): p. 438-448. 92. Wang, D.-H., et al., Reduced serum levels of ALT and GGT and high carbohydrate intake among workers exposed to toluene below the threshold limit values. Industrial Health, 1998. 36(1): p. 14-19.

93. Paez-Martinez, N., et al., Participation of GABAA, GABAB receptors and neurosteroids in toluene-induced hypothermia: Evidence of concentration-dependent differences in the mechanism of action. European Journal of Pharmacology, 2013. 698(1-3): p. 178-185. 94. Connors, N.J. Inhalants Clinical Presentation. 2017 [cited 2018 20 March 2018]; Available from: https://emedicine.medscape.com/article/1174630-clinical. 95. Hales, C.N. and D.J. Barker, The thrifty phenotype hypothesis. British Medical Bulletin, 2001. 60(1): p. 5-20. 96. DiMagno, E.P., V.L. Go, and W. Summerskill, Relations between pancreatic enzyme outputs and malabsorption in severe pancreatic insufficiency. New England Journal of Medicine, 1973. 288(16): p. 813-815. 97. Feitelberg, S., et al., Deranged bone mineral metabolism in chronic alcoholism. Metabolism, 1987. 36(4): p. 322-326. 98. Diamond, T., et al., Ethanol reduces bone formation and may cause osteoporosis. The American Journal of Medicine, 1989. 86(3): p. 282-288. 99. Lindtner, C., et al., induces whole-body insulin resistance by impairing hypothalamic insulin action. Science Translational Medicine, 2013. 5(170): p. 170ra14.

100. Beckstead, M.J., et al., Glycine and γ-Aminobutyric AcidA Receptor Function Is Enhanced By Inhaled Drugs Of Abuse. Molecular Pharmacology, 2000. 57(6): p. 1199-1205. 101. Cruz, S.L., et al., Effects of the Abused Solvent Toluene on Recombinant N-Methyl-d- Aspartate and non-N-Methyl-d-Aspartate Receptors Expressed inXenopus Oocytes. Journal of Pharmacology and Experimental Therapeutics, 1998. 286(1): p. 334-340. 102. Brumberg, J.J., Fasting girls: The emergence of anorexia nervosa as a modern disease. 1988, Cambridge, MA: Harvard University Press. 103. Gordon, C.M. The impact of anorexia nervosa on bone health. in International Congress Series. 2007. Elsevier. 104. Soyka, L.A., et al., The Effects of Anorexia Nervosa on Bone Metabolism in Female Adolescents 1. The Journal of Clinical Endocrinology & Metabolism, 1999. 84(12): p. 4489-4496.

142 Rose Crossin (737900)

105. Grinspoon, S., et al., Serum leptin levels in women with anorexia nervosa. The Journal of Clinical Endocrinology & Metabolism, 1996. 81(11): p. 3861-3863. 106. Hartman, D., et al., Bone density of women who have recovered from anorexia nervosa. International Journal of Eating Disorders, 2000. 28(1): p. 107-112. 107. Valla, Å., et al., Anorexia nervosa: slow regain of bone mass. Osteoporosis International, 2000. 11(2): p. 141-145. 108. Atay, A.A., et al., Bone mass toxicity associated with inhalation exposure to toluene. Biological Trace Element Research, 2005. 105(1-3): p. 197-203. 109. Cornish, J., et al., Leptin directly regulates bone cell function in vitro and reduces bone fragility in vivo. Journal of Endocrinology, 2002. 175(2): p. 405-415. 110. Cornish, J., et al., Systemic administration of amylin increases bone mass, linear growth, and adiposity in adult male mice. American Journal of Physiology- Endocrinology And Metabolism, 1998. 275(4): p. E694-E699. 111. Haffner, S.M. and R.L. Bauer, The association of obesity and glucose and insulin concentrations with bone density in premenopausal and postmenopausal women. Metabolism, 1993. 42(6): p. 735-738. 112. Won, Y.L., et al., The Effects of Long-Term, Low-Level Exposure to Monocyclic Aromatic Hydrocarbons on Worker's Insulin Resistance. Safety and Health at Work, 2011. 2(4): p. 365-374. 113. Hwang, Y.-C., et al., Circulating osteocalcin level is associated with improved glucose tolerance, insulin secretion and sensitivity independent of the plasma adiponectin level. Osteoporosis International, 2012. 23(4): p. 1337-1342. 114. Ferron, M., et al., Osteocalcin differentially regulates β cell and adipocyte gene expression and affects the development of metabolic diseases in wild-type mice. Proceedings of the National Academy of Sciences, 2008. 105(13): p. 5266-5270. 115. Lee, N.K. and G. Karsenty, Reciprocal regulation of bone and energy metabolism. Trends in Endocrinology & Metabolism, 2008. 19(5): p. 161-166. 116. Fukumoto, S. and T.J. Martin, Bone as an endocrine organ. Trends in Endocrinology & Metabolism, 2009. 20(5): p. 230-236. 117. DiGirolamo, D.J., T.L. Clemens, and S. Kousteni, The skeleton as an endocrine organ. Nature Reviews Rheumatology, 2012. 8(11): p. 674-683. 118. Karsenty, G., Convergence between bone and energy homeostases: leptin regulation of bone mass. Cell Metabolism, 2006. 4(5): p. 341-348. 119. Hsieh, G., R. Sharma, and R. Parker, Hypothalamic-pituitary-adrenocortical axis activity and immune function after oral exposure to benzene and toluene. Immunopharmacology, 1991. 21(1): p. 23-32. 120. Bowen, S.E., et al., Abstinence following toluene exposure increases anxiety-like behavior in mice. Neurotoxicology and Teratology, 2017. 121. Paez-Martinez, N., et al., Environmental enrichment increases doublecortin-associated new neurons and decreases neuronal death without modifying anxiety-like behavior in mice chronically exposed to toluene. Behavioural Brain Research, 2013. 256: p. 432- 440. 122. Hougaard, K.S., et al., Toluene depresses plasma corticosterone in pregnant rats. Basic & Clinical Pharmacology & Toxicology, 2003. 92(3): p. 148-152. 123. Bowen, S.E., J.L. Wiley, and R.L. Balster, The effects of abused inhalants on mouse behavior in an elevated plus-maze. European Journal of Pharmacology, 1996. 312(2): p. 131-136. 124. Harvey, P.W., D.J. Everett, and C.J. Springall, Adrenal toxicology: a strategy for assessment of functional toxicity to the adrenal cortex and steroidogenesis. Journal of Applied Toxicology, 2007. 27(2): p. 103-115.

143 Rose Crossin (737900)

125. Verma, Y. and S.V.S. Rana, Endocrinal toxicity of industrial solvents–a mini review. Indian Journal of Experimental Biology, 2009. 47(7): p. 537-549. 126. Yang, M., et al., Toluene Induces Depression-Like Behaviors in Adult Mice. Toxicological Research, 2010. 26(4): p. 315. 127. Gotohda, T., I. Tokunaga, and S.-i. Kubo, Toluene inhalation-induced adrenocortical hypertrophy and endocrinological changes in rat. Life Sciences, 2005. 76(17): p. 1929- 1937. 128. Kamijo, Y., et al., Fatal bilateral adrenal hemorrhage following acute toluene poisoning: a case report. Journal of Toxicology: Clinical Toxicology, 1998. 36(4): p. 365- 368. 129. Harvey, P.W. and C. Sutcliffe, Adrenocortical hypertrophy: establishing cause and toxicological significance. Journal of Applied Toxicology, 2010. 30(7): p. 617-626. 130. Arlt, W. and B. Allolio, Adrenal insufficiency. The Lancet, 2003. 361(9372): p. 1881- 1893. 131. Canadian Paediatric Society, F.N., Inuit and M.H.C.B. L, Inhalant abuse. Paediatrics and Child Health, 2010. 15(7): p. 443-448. 132. Kretsch, M., et al., Cognitive effects of a long-term weight reducing diet. International Journal of Obesity, 1997. 21(1): p. 14-21. 133. Sharp, C. and C. Freeman, The medical complications of anorexia nervosa. The British Journal of Psychiatry, 1993. 162(4): p. 452-462. 134. Molinari, E., et al., Psychological and emotional development, intellectual capabilities, and body image in short normal children. Journal of Endocrinological Investigation, 2002. 25(4): p. 321-328. 135. Siegel, P., R. Clopper, and B. Stabler, Psychological impact of significantly short stature. Acta Paediatrica Scandinavica. Supplement, 1991. 377: p. 14-18. 136. Dulloo, A.G., et al., The thrifty ‘catch-up fat’phenotype: its impact on insulin sensitivity during growth trajectories to obesity and metabolic syndrome. International Journal of Obesity, 2006. 30: p. S23-S35. 137. Virmani, A., et al., Links between nutrition, drug abuse, and the metabolic syndrome. Annals of the New York Academy of Sciences, 2006. 1074(1): p. 303-314. 138. Cowan, J.A. and C.M. Devine, Process evaluation of an environmental and educational nutrition intervention in residential drug-treatment facilities. Public Health Nutrition, 2012. 15(07): p. 1159-1167. 139. Zoli, M. and M.R. Picciotto, Nicotinic regulation of energy homeostasis. Nicotine & Tobacco Research, 2012. 14(11): p. 1270-1290. 140. Penner, E., H. Buettner, and M. Mittleman, Recent use of marijuana affects insulin resistance. American Journal of Medicine, 2013. 141. Nebhinani, N., et al., Prevalence of the metabolic syndrome in substance-dependent men. German Journal of Psychiatry, 2013. 16(2): p. 61-67. 142. Nolan, L.J., Shared urges? The links between drugs of abuse, eating, and body weight. Current Obesity Reports, 2013. 2(2): p. 150-156. 143. Crossin, R., et al., Altered body weight associated with substance abuse: a look beyond food intake. Addiction Research & Theory, 2018: p. 1-9. 144. Burns, C., P. d'Abbs, and B. Currie, Patterns of petrol sniffing and other drug use in young men from an Australian Aboriginal community in Arnhem Land, Northern Territory. Drug and Alcohol Review, 1995. 14(2): p. 159-169. 145. Wallen, M., P. Näslund, and M.B. Nordqvist, The effects of ethanol on the kinetics of toluene in man. Toxicology and Applied Pharmacology, 1984. 76(3): p. 414-419. 146. White, A.M., et al., Differential effects of ethanol on motor coordination in adolescent and adult rats. Pharmacology Biochemistry and Behavior, 2002. 73(3): p. 673-677.

144 Rose Crossin (737900)

147. Chambers, R.A., J.R. Taylor, and M.N. Potenza, Developmental neurocircuitry of motivation in adolescence: a critical period of addiction vulnerability. American Journal of Psychiatry, 2014. 148. MacPhail, R., J. Farmer, and K. Jarema, Toluene effects on the motor activity of adolescent, young-adult, middle-age and senescent male Brown Norway rats. Neurotoxicology, 2012. 33(1): p. 111-118. 149. Lubman, D.I., M. Yücel, and W.D. Hall, Substance use and the adolescent brain: a toxic combination? Journal of Psychopharmacology, 2007. 21(8): p. 792-794. 150. Samuel‐Herter, S.R., S.L. Slaght, and B.E. McKay, Age‐dependent time courses of recovery for motor functions following acute toluene intoxication in rats. Developmental Psychobiology, 2014. 56(4): p. 657-673. 151. Cairney, S., et al., Neurological and cognitive recovery following abstinence from petrol sniffing. Neuropsychopharmacology, 2005. 30(5): p. 1019-1027. 152. Cairney, S., et al., A prospective study of neurocognitive changes 15 years after chronic inhalant abuse. Addiction, 2013. 108(6): p. 1107-1114. 153. Benignus, V.A., et al., Quantitative comparisons of the acute neurotoxicity of toluene in rats and humans. Toxicological Sciences, 2007. 100(1): p. 146-155. 154. Mehlem, A., et al., Imaging of neutral lipids by oil red O for analyzing the metabolic status in health and disease. Nature Protocols, 2013. 8(6): p. 1149-1154. 155. Uchoa, E.T., et al., Glucocorticoids are required for meal-induced changes in the expression of hypothalamic neuropeptides. Neuropeptides, 2012. 46(3): p. 119-124. 156. Akana, S.F., et al., Corticosterone: narrow range required for normal body and thymus weight and ACTH. American Journal of Physiology-Regulatory, Integrative and Comparative Physiology, 1985. 249(5): p. R527-R532. 157. Northern Territory Department of Health and Families, Child Malnutrition / Failure to Thrive. 2008, Northern Territory Department of Health and Families: Alice Springs. 158. Perrin, E.C., et al., Criteria for Determining Disability in Infants and Children: Failure to Thrive: Summary. 2003. 159. Corbett, S. and R. Drewett, To what extent is failure to thrive in infancy associated with poorer cognitive development? A review and meta‐analysis. Journal of Child Psychology and Psychiatry, 2004. 45(3): p. 641-654. 160. Maggioni, A. and F. Lifshitz, Nutritional management of failure to thrive. Pediatric Clinics of North America, 1995. 42(4): p. 791-810. 161. Norgan, N., Long-term physiological and economic consequences of growth retardation in children and adolescents. Proceedings of the Nutrition Society, 2000. 59(02): p. 245- 256. 162. Lubman, D.I., L. Hides, and M. Yucel, Inhalant misuse in youth: time for a coordinated response. Medical Journal of Australia, 2006. 185(6): p. 327. 163. Olgar, S., et al., Volatile solvent abuse caused glomerulopathy and tubulopathy in street children. Human & Experimental Toxicology, 2008. 27(6): p. 477-483. 164. Uchino, A., et al., Comparison between patient characteristics and cranial MR findings in chronic thinner intoxication. European radiology, 2002. 12(6): p. 1338-1341. 165. Frisch, R.E., Fatness, menarche, and female fertility. Perspectives in Biology and Medicine, 1985. 28(4): p. 611-633. 166. Frisch, R.E., R. Revelle, and S. Cook, Components of weight at menarche and the initiation of the adolescent growth spurt in girls: estimated total water, lean body weight and fat. Human Biology, 1973: p. 469-483. 167. Burns, C.B., et al., Evaluation of strategies used by a remote aboriginal community to eliminate petrol sniffing. The Medical Journal of Australia, 1995. 163(2): p. 82-86. 168. Mellor, D., et al., Body image importance and body dissatisfaction among Indigenous Australian adolescents. Body Image, 2004. 1(3): p. 289-297.

145 Rose Crossin (737900)

169. Greenberg, I., et al., Effects of marihuana use on body weight and caloric intake in humans. Psychopharmacology, 1976. 49(1): p. 79-84. 170. Wannamethee, S.G. and A.G. Shaper, Alcohol, body weight, and weight gain in middle- aged men. The American Journal of Clinical Nutrition, 2003. 77(5): p. 1312-1317. 171. Compston, J., et al., Effect of diet-induced weight loss on total body bone mass. Clinical Science, 1992. 82(4): p. 429-432. 172. Misra, M., et al., Alterations in cortisol secretory dynamics in adolescent girls with anorexia nervosa and effects on bone metabolism. The Journal of Clinical Endocrinology & Metabolism, 2004. 89(10): p. 4972-4980. 173. Leibel, R.L., M. Rosenbaum, and J. Hirsch, Changes in energy expenditure resulting from altered body weight. New England Journal of Medicine, 1995. 332(10): p. 621- 628. 174. Cohen, S., et al., Chronic stress, glucocorticoid receptor resistance, inflammation, and disease risk. Proceedings of the National Academy of Sciences, 2012. 109(16): p. 5995- 5999. 175. Melhem, N.M., et al., Blunted HPA axis activity in suicide attempters compared to those at high risk for suicidal behavior. Neuropsychopharmacology, 2016. 41(6): p. 1447. 176. O’Connor, D.B., et al., Cortisol reactivity and suicidal behavior: investigating the role of hypothalamic-pituitary-adrenal axis responses to stress in suicide attempters and ideators. Psychoneuroendocrinology, 2017. 75: p. 183-191. 177. Han, Z., et al., Maternal underweight and the risk of preterm birth and low birth weight: a systematic review and meta-analyses. International Journal of Epidemiology, 2010. 40(1): p. 65-101. 178. Harms, M. and P. Seale, Brown and beige fat: development, function and therapeutic potential. Nature Medicine, 2013. 19(10): p. 1252-1263. 179. Fries, E., et al., A new view on hypocortisolism. Psychoneuroendocrinology, 2005. 30(10): p. 1010-1016. 180. Buckingham, J.C. and J. Hodges, Interrelationships of pituitary and plasma corticotrophin and plasma corticosterone in adrenalectomized and stressed, adrenalectomized rats. Journal of Endocrinology, 1974. 63(1): p. 213-222. 181. Hodges, J. and J. Vernikos-Danellis, Pituitary and blood corticotrophin changes in adrenalectomized rats maintained on physiological doses of . Acta Endocrinologica, 1962. 39(1): p. 79-86. 182. Adler, N.E., et al., Socioeconomic status and health: the challenge of the gradient. American Psychologist, 1994. 49(1): p. 15.

146 Rose Crossin (737900)

Appendix 1 – Review on the effects of substance abuse on body weight This research highlighted that inhalant abuse can have a profound effect on body weight, but that food intake is not the sole mediator of these changes. Many other substances of abuse are associated with changes to body weight, and the predominant hypothesis in the substance abuse field is that this is due to changes to food intake, without consideration of the rest of the energy balance equation. Therefore, a think-piece was published, aiming to consider potential causes of body weight changes during active substance abuse and abstinence, by focussing on the components of the energy balance equation. This review was published as: Altered body weight associated with substance abuse: a look beyond food intake.

Citation:

Crossin, R., Lawrence, A. J., Andrews, Z. B., & Duncan, J. R. (2018). Altered body weight associated with substance abuse: a look beyond food intake. Addiction Research & Theory, 1- 9.

147 ADDICTION RESEARCH & THEORY, 2018 https://doi.org/10.1080/16066359.2018.1453064

VIEWPOINT Altered body weight associated with substance abuse: a look beyond food intake

Rose Crossina,b , Andrew J. Lawrencea,c, Zane B. Andrewsd and Jhodie R. Duncana,e aAddiction Neuroscience, Florey Institute of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia; bEastern Health Clinical School, Monash University, Box Hill, VIC, Australia; cFlorey Department of Neuroscience and Mental Health, University of Melbourne, Melbourne, VIC, Australia; dMetabolic Disease and Obesity Theme, Biomedicine Discovery Institute, Department of Physiology, Monash University, Clayton, Victoria, Australia; eSchool of Medicine, University of Adelaide, Adelaide, SA, Australia

ABSTRACT ARTICLE HISTORY Background: Substance abuse can cause a range of harmful secondary health consequences, including Received 24 September 2017 body weight changes. These remain poorly understood but can lead to metabolic disorders including Revised 5 March 2018 obesity and diabetes. Energy balance is a function of the equation: energy balance ¼ energy intake – Accepted 12 March 2018 energy expenditure; an imbalance to this equation results in body weight changes. Currently, in the KEYWORDS clinical setting, changes to food intake (energy intake) are considered as the primary mediator of body Energy balance equation; weight changes related to substance abuse, reflected in the current treatment focus on nutritional pair-feeding; abstinence; intervention. The influence of substance abuse on energy expenditure receives less attention. The aim energy expenditure; of this think-piece is to consider potential causes of body weight changes during active substance nutrition; thermogenesis abuse and abstinence, by focussing on the components of the energy balance equation. Methods: We discuss both human and animal studies on the effects of substance abuse on energy bal- ance, with particular focus on animal models utilising pair-feeding, which enable investigation of energy balance whilst controlling for the effects of altered food intake. Results: We demonstrate that whilst some drugs of abuse affect food intake, this effect is inconsistent. Furthermore, body weight changes do not match food intake changes. Conclusion: We provide evidence that drugs of abuse can affect both energy intake and energy expenditure; contributing to the observed body weight changes. This think-piece highlights that treat- ment strategies for body weight changes related to substance abuse cannot focus solely on nutritional interventions, but should consider the impact of broader disruptions to energy balance.

Introduction however, the secondary health consequences receive less focus and there are substantial knowledge gaps. A major sec- Substance abuse is a widespread and pressing public health ondary health consequence that remains poorly understood issue worldwide (United Nations Office of Drugs and Crime is the effect of substance abuse on body weight. Active sub- 2014; Naghavi et al. 2015). Included within substance abuse stance abuse has been associated with decreased body weight are illegal substances such as cocaine, heroin or metham- (Cofrancesco et al. 2007) and many individuals report phetamine, and legal substances such as alcohol and ciga- unhealthy weight gain after drug use ceases in recovery/ rettes. Both substance abuse and its health consequences abstinence (Hodgkins et al. 2004; Cowan and Devine 2008). ’ significantly reduce the quality of an individual s life and This would suggest that drugs of abuse may affect an indi- increase the risk of drug-related illness and injury. As such, vidual’s energy balance. Dramatic changes to body weight, substance abuse is considered a non-communicable disease; both decreases and increases, can affect an individual’s long- collectively responsible for approximately two-thirds of the term health and contribute to the onset of cardiovascular world’s deaths (World Health Organisation 2014). diseases and metabolic diseases such as diabetes (Dulloo Substance abuse also accounts for one of the largest et al. 2006). health costs to society, in Australia alone for example, the The energy balance equation is: energy balance ¼ energy health costs associated with substance abuse are estimated as intake - energy expenditure, with our body weight a function being over $2.5 billion per year (Collins and Lapsley 2008). of the energy stored in our body. Thus, an imbalance in this Of these health costs, a large proportion are not from the equation can lead to changes in body weight. Energy intake direct effects of drug intoxication, but from secondary health is primarily driven by food intake and moderated by impacts, such as cancer or stroke (Collins and Lapsley 2008). absorption. Energy expenditure includes basal metabolic rate, Researchers and clinicians place a large focus on understand- physical activity and thermogenesis (subdivided into ing the underlying causes leading to substance abuse, cold-induced thermogenesis and the thermic effect of food).

CONTACT Rose Crossin [email protected] Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria 3010, Australia ß 2018 Informa UK Limited, trading as Taylor & Francis Group 2 R. CROSSIN ET AL.

This equation is controlled by the processes of metabolism, The effects of substance abuse on body weight which encompasses a system that regulates eating behaviour, nutrient absorption, as well as the storage and release of To start to understand the overall relationship between sub- energy. Body weight will remain stable when there is a bal- stance abuse and body weight, it is helpful to first separate active substance abuse from abstinence (i.e. post-drug use), ance between energy intake, storage, and expenditure, known as these can result in very different effects on body weight. as energy homeostasis. The maintenance of body weight is Despite the lack of consensus in human studies (Nolan 2013; buffered, i.e. a decrease in energy intake results in decreased Sansone and Sansone 2013; Meule 2014), in animal models expenditure and vice versa, therefore, any changes in body active substance abuse is more commonly associated with weight occur by overwhelming the buffering potential of low body weight in comparison to controls for many drug homeostatic systems (Keesey and Powley 2008). A negative types including opioids (Thornhill et al. 1976), cocaine energy balance occurs when energy intake is less than energy (Planeta and Marin 2002), nicotine (Grebenstein et al. 2013), expended, and this shortfall is made up by a release of methamphetamine (Williams et al. 2004), marijuana energy from body stores, leading to reduced body weight. If (Rahminiwati and Nishimura 1999; Ignatowska-Jankowska the difference between energy intake and energy expenditure et al. 2011), and 3,4-methylenedioxymethamphetamine is positive, then energy is stored and body weight increases (MDMA) (Piper et al. 2005). However, findings on body (positive energy balance). Physiological processes strongly weight are inconsistent in animal models of alcohol con- defend body weight decreases arising from a negative energy sumption (Larue-Achagiotis et al. 1990; Vetter et al. 2007). balance, ultimately leading to weight regain (Sumithran and In clinical studies, the predominant hypothesis explaining Proietto 2013). In contrast, a positive energy balance that weight loss during active substance abuse is that appetite is leads to weight gain does not initiate the same physiological suppressed (Santolaria-Fernandez et al. 1995; Islam et al. defence, and weight gain can continue into obesity, and the 2002), and that food intake is reduced due to a loss of inter- new elevated body weight is defended (Keesey 1989). est in meals (Santolaria et al. 2000) or poor eating habits Despite the persistent trope of the emaciated person with (Neale et al. 2012; Petersson et al. 2004). This has led to a a drug addiction in popular culture (Linnemann and Wall conclusion that active substance abuse results in a negative 2013), there remains no clear scientific consensus on the energy balance and therefore loss of body weight (Mohs effects of substance abuse on body weight in humans, both et al. 1990) via a reduction in energy intake. during active substance abuse and in abstinence (Nolan In contrast to active substance abuse, recovery/abstinence 2013; Sansone and Sansone 2013; Meule 2014), which is a from substance abuse has a strong association with rapid significant gap in our understanding of the health conse- weight gain leading to a body mass index (BMI) above the quences of substance abuse. There are various theories in the normal range, which occurs across genders and age groups, literature to suggest how substance abuse may result in and in relation to abstinence from alcohol, cigarettes, and altered body weight in humans, however, the common elem- illicit drugs (Jackson and Grilo 2002; Hodgkins et al. 2004). ent of these is that they focus on food intake (energy intake) It is not always clear unfortunately, whether this represents as the primary driver of body weight changes. The other weight normalisation (after a period of weight loss) or exces- components of the energy balance equation, e.g. energy sive weight gain. However, self-reports from individuals expenditure or nutrient absorption, receive less focus when recovering from substance abuse suggest that weight gain is considering the relationship between substance abuse and excessive (Cowan and Devine 2008). Studies of patients in body weight. This is despite a quantity of knowledge from recovery from substance abuse report increases in body animal models, which show substance abuse can affect weight between 3.4 and 27.2 kg, which occurs predominantly energy expenditure as well as energy intake. at the start of the recovery period (Nolan and Stolze 2012). This think-piece article discusses body weight changes This is such a well-reported occurrence that an individual’s associated with substance abuse, and the potential causes of concerns about increasing body weight can be a barrier to these changes, with a focus on the various components of commencing treatment for substance abuse (Warren the energy balance equation. Particular emphasis will be et al. 2013). given to animal studies utilising pair-feeding, in which body When considering the causes of observed weight gain weight changes can be investigated whilst controlling for the during recovery from substance abuse there are three main effects of altered food intake, and to animal studies that theories, though all can be challenged: measure energy expenditure as well as input. Understanding how substance abuse can affect body weight will help under- stand how drugs of abuse act within the body, the long-term 1. Rebound effect - given the association between low health risks for individuals with a history of substance abuse, body weight and active substance abuse, the weight and could guide treatment strategies. If food intake is the increase during abstinence may be attributed to a sole mediator of body weight changes related to substance rebound effect, as homeostatic mechanisms seek to nor- abuse, then nutritional interventions should be able to coun- malise body weight (Nolan 2013). However, the increase teract these body weight changes. However, if substance in food intake during abstinence is in excess of that abuse causes disruptions to energy balance beyond food needed to return to a normal body weight (Cowan and intake, then additional treatment strategies will need to be Devine 2008) and food choices during recovery may be developed with these considerations in mind and translated driven more by the rewarding values of food than by into clinical practice. nutritional quality (Cowan and Devine 2008) implying ADDICTION RESEARCH & THEORY 3

that hedonic mechanisms may override homeo- abuse, then it follows that for any drug, which is associated static mechanisms. with decreased body weight, it should also be associated with 2. ‘Addiction switching’ to palatable foods - weight gain decreased food intake. However, this is not the case, and may be attributable to an increased consumption of whilst some drugs of abuse are associated with decreased highly palatable foods, which are consumed as a replace- food intake, not all are. Decreased food intake is commonly ment for the drug – a model of ‘addiction switching’ observed in relation to use of methamphetamine (Honma from a drug to food (Nolan 2013; Brunault et al. 2015). and Honma 2009), heroin (Thornhill et al. 1976) and However, this does not explain why patients in metha- MDMA (Freudenmann et al. 2006). Whilst these drugs do done treatment rapidly gain weight, because there is a suppress appetite and decrease food intake, and therefore still a rewarding substance in the brain, which could have the potential to result in decreased body weight, it is theoretically displace palatable foods (Gambera and unclear if the change in body weight is proportional to the Clarke 1976; Mysels et al. 2011), despite the lack of pro- change in food intake, or if there are other concurrent effects cedural and/or social rewards. This would imply, in con- on energy balance. trast to the ‘rebound effect’ above, that hedonic or In contrast, appetite suppression is not a consistent fea- reward mechanisms are not the sole driver of this effect. ture of chronic cigarette smoking (Natividad et al. 2013)or 3. Reward seeking phenotype - both substance abuse and cocaine use, yet long-term use of these drugs is associated increased body weight caused by increased food con- with body weight decreases (Planeta and Marin 2002; sumption in abstinence may be outcomes of a common Grebenstein et al. 2013). In human studies, smokers typically behavioural tendency towards reward seeking or impul- eat the same amount or slightly more than non-smokers, sivity (Nolan and Stolze 2012). However, if the sole rela- which does not match with the observed weight suppression tionship between drugs of abuse and food intake was effects (Perkins 1992). Cocaine also has a reputation for such that they derived from a common phenotype of appetite suppression to the extent that some individuals reward seeking, then a naïve individual with no previous report this is a primary driver for their use of the drug knowledge of the relative rewards of food or drugs (Cochrane et al. 1998), though this is challenged by animal should have an equal preference for them both. In studies that show that acute exposure to cocaine delays, but humans, this is difficult to investigate due to prior does not reduce food intake and that the anorectic effects are transitory (Balopole et al. 1979; Cooper and Van der exposure to rewarding food, but it can be tested in ani- Hoek 1993). Therefore, for some drugs of abuse, appetite mal models using choice experiments, and these types of suppressing effects are absent, inconsistent, or do not match studies suggest that food rewards are predominantly with the observed effects on body weight (Balopole et al. chosen over drugs (Lenoir et al. 2007; LeSage 2009; 1979; Perkins 1992; Cooper and Van der Hoek 1993; Caprioli et al. 2015). Natividad et al. 2013). This would suggest that the effects of these drugs on body weight are not solely due to The focus on food intake (energy intake) as the mediator food intake. of either decreased body weight during active substance abuse or increased body weight in abstinence logically leads to a conclusion that nutritional interventions, in order to The effects of substance abuse on body weight normalise food intake, will then normalise body weight independent of food intake (Cowan and Devine 2012). Indeed, nutritional interventions In humans, it could be argued that the inconsistencies in the remain a key treatment strategy in the clinical environment relationship between food intake and body weight could be (Australian Drug Foundation 2016). We hypothesise that due to other factors, such as poly-drug use or socio-eco- whilst substance abuse may alter food intake, substance nomic factors. However animal studies, particularly those abuse may also affect other components of the energy bal- that utilise pair-feeding, can elucidate this relationship more ance equation independently of food intake, thus contribu- clearly. In a pair-feeding protocol, drug-free animals only ting to weight loss during active substance abuse and weight have access to the same amount of food as is consumed by gain in abstinence. Whilst disruptions to energy balance the drug-exposed animals, which enables the direct effect of independent of food intake have been demonstrated in ani- the drug to be separated from the secondary effect of altered mal models, translation to human studies and clinical prac- food intake. Many pair-feeding animal studies, which will be tice is limited, and thus the health consequences remain discussed below, reveal that even for drugs where it appears unknown. If left untreated, energy balance disruptions have that there is a relationship between food intake and weight, the potential to cause long-lasting health impacts for individ- whether positive or negative, the nature of this relationship uals, which persist even if drug use ceases (Dulloo is more complex than a simple change to energy balance et al. 2006). induced by altered food intake. Whilst a pair-feeding proto- col controls for the effects of altered food intake between The effects of active substance abuse on groups, this type of study is unfortunately not common. food intake However, the hypothesis that body weight changes in rela- tion to substance abuse are not solely attributable to food If the current hypotheses are correct, and food intake is the intake can also be supported by studies measuring energy sole driver of body weight changes in relation to substance expenditure, even without a pair-feeding protocol. 4 R. CROSSIN ET AL.

Decreased food intake with decreased body weight Kubler et al. 2013). Therefore, opioids have a positive effect relative to food intake (methamphetamine, MDMA, on body weight in relation to food intake, and body weight and cigarettes) is higher than what can be accounted for by food intake alone (Pinter-Kubler et al. 2013). Potential causes of this Whilst food intake is reduced in response to chronic expos- change to the relationship between food intake and body ure to injected methamphetamine (2 mg/kg) in adult male weight in -exposed animals include decreased energy rats, pair-feeding demonstrated that weight loss is more expenditure due to hypo-locomotion (Kauppila et al. 1992) severe than what is attributable to altered food intake (Caul or reduced gut motility, which affects food absorption et al. 1988). Similarly, injected MDMA (10 mg/kg) adminis- (Williams et al. 1997). tered to adult male rats rapidly reduces both food intake and body weight (Francis et al. 2011), though unfortunately we could not find any pair-feeding studies to probe this rela- Unchanged food intake with decreased body weight tionship. In mouse studies of chronic cigarette smoke expos- relative to food intake (alcohol and cocaine) ure via inhalation, weight impairment in exposed male animals is greater than what can be accounted for by food Chronic exposure to injected cocaine (10-30 mg/kg) in male intake (Chen et al. 2006). The latter study was conducted in rats has not been associated with changes to daily food adolescent mice, when body weight is still increasing, and intake, despite short-term disruptions to feeding behaviour whilst the rate of increase was reduced in both exposed and immediately following exposure (Balopole et al. 1979; pair-fed animals, the body weight impairment occurred Cooper and Van der Hoek 1993), though it is associated faster in the exposed animals (Chen et al. 2006). with reductions in body weight (Planeta and Marin 2002). Furthermore, adiposity was only reduced in those animals Similarly, chronic alcohol consumption (20% ethanol in exposed to cigarette smoke, suggesting that broader changes water as sole liquid available) is not associated with food to energy balance were occurring, in addition to reduced intake changes in male rats, however, body weight gain is food intake (Chen et al. 2006). These findings would suggest impaired (Larue-Achagiotis et al. 1990), with the authors that additional effects on energy balance are occurring fol- suggesting that energy expenditure had increased, potentially lowing methamphetamine, MDMA, and cigarette in response to increased alcohol metabolism. smoke exposure. Unfortunately, no pair-feeding studies on the effects of Though all of the above substances can increase energy cocaine on body weight in adolescent or adult animal mod- expenditure, which may explain the observed effects on body els could be found, however one human study that seems to weight, the component of energy expenditure that is affected resolve this paradox showed that is asso- varies between substances. Medium and high doses of ciated with significantly higher intake of fat and carbohy- injected methamphetamine (0.3–10 mg/kg) are associated drates and binge eating behaviour, but body weight is with hyper-locomotion (Singh et al. 2012) and hyperthermia reduced due to impaired fat regulation (Ersche et al. 2013). (Makisumi et al. 1998), whereas MDMA is also associated When considering the effects of alcohol on body weight, with hyper-locomotion (when injected at 10 mg/kg) (Francis care must be taken given that alcohol is caloric and can et al. 2011), but also increased basal metabolic rate (ingested therefore be a source of energy intake. However, pair-feeding 2 mg/kg) (Freedman et al. 2005). Additionally, MDMA is studies have shown that alcohol can induce a negative also associated with increased urination and defaecation energy balance independently of food intake, though this (when injected at doses of both 6.3 and 20 mg/kg) (Bilsky effect was only observed when alcohol was injected (0.3 g/ et al. 1991), which may additionally affect energy balance via 100 g body weight) rather than administered by gavage at decreased food absorption. In relation to cigarette smoking, the same dose (Luz et al. 1996). This may be because the there is also evidence of increased energy expenditure effect of the calories consumed from gavaged alcohol may (Hofstetter et al. 1986; Audrain-McGovern and Benowitz counteract energy expenditure, whereas the calories from 2011), increased thermogenesis (Lupien and Bray 1988), and alcohol would not be relevant using an injection method. an altered inflammatory profile leading to cachexia (sickness anorexia) (Chen et al. 2008). These studies highlight that Increased food intake with inconsistent effects on body even when substances have a similar effect on food intake weight relative to food intake (marijuana) and body weight, the mode of effect is substance-specific. Marijuana is associated with increased food intake in Decreased food intake with increased body weight humans (Greenberg et al. 1976; Foltin et al. 1986; Rodondi relative to food intake (opioids) et al. 2006). In some human studies, the subsequent increase in body weight associated with this increased food intake is Opioid administration by injection over 7 days can reduce greater than predicted by caloric intake alone (Greenberg daily food consumption by a third in adult male rats et al. 1976; Foltin et al. 1988), whereas others find that whilst (Thornhill et al. 1976). However, whilst there is an observed food intake is increased, BMI does not increase (Rodondi decrease in body weight after 7 days of opioid administra- et al. 2006). In a study using adolescent male mice, chronic tion, the body weight loss caused by opioids is less than exposure to marijuana (injected D9-Tetrahydrocannabinol) what occurs in pair-fed controls, suggesting a decrease in the resulted in no changes to food intake, though higher doses metabolic activity of animals exposed to opioids (Pinter- were associated with reduced weight gain (Rahminiwati and ADDICTION RESEARCH & THEORY 5

Table 1. Grouping drugs of abuse by their effect on food intake and by their effect on body weight relative to that food intake. Effect on Effect on body weight Drug of abuse food intake relative to food intake Hypo-/hyper-metabolic Methamphetamine Decreased Decreased Hyper-metabolic MDMA Decreased Decreased Hyper-metabolic Cigarettes Decreased Decreased Hyper-metabolic Opioids Decreased Increased Hypo-metabolic Alcohol Unchanged Decreased Hyper-metabolic Cocaine Unchanged Decreased Hyper-metabolic Marijuana Increased Inconsistent findings Unknown This table groups different drugs of abuse first by their effect on food intake and second by their effects on body weight relative to that food intake, during active substance abuse. The effects of marijuana are inconsistent between studies; hence, no hypo-/hyper-metabolic grouping has been made for this drug. Marijuana has been shown in increase both energy intake and expenditure, and these competing effects may be partially confounded by water retention affecting body weight. Whilst in pair-feeding studies cigarettes have a neutral effect on body weight, it should be noted that the weight loss occurs faster in exposed animals than in pair-fed and there are other associated metabolic and body composition changes that occur independently of food intake, thus cigarettes are grouped as being hyper-metabolic.

Nishimura 1999) suggestive of an increase in energy expend- food intake. This is shown particularly clearly in pair-feeding iture. Indeed, in animal models, marijuana may have a non- studies, though it should be noted that pair-feeding protocols linear relationship on food intake, with increased food intake only take into account total daily calories consumed, they do observed at lower doses and decreased food intake observed not take into account changes to eating habits or patterns of at higher doses, at least in males (Wiley et al. 2005), which how those calories are consumed. Using these studies, drugs will then influence the effect on body weight. This relation- of abuse can be grouped by first delineating their effects on ship may also be influenced by the frequency of food intake food intake, and secondly how body weight changes in rela- measures, as marijuana can induce acute hyperphagia within tion to that food intake, as seen in Table 1. For alcohol, the first hours following exposure, without a change to total cocaine, cigarettes, methamphetamine and MDMA, this daily food intake (Williams et al. 1998). effect may be described as hyper-metabolic, whereby body There are two potential causes for the inconsistencies weight changes are negative in relation to food intake, and observed in this relationship. Firstly, smoked marijuana can body weight is lower than what can be accounted for by food increase metabolic rate, and thus energy expenditure intake alone. For opioids, this effect may be described as hypo- (Zwillich et al. 1978). This increase in energy expenditure metabolic, whereby body weight changes are positive (or neu- may counteract the increase in energy intake; however, the tral) in relation to food intake. Marijuana is difficult to charac- effect on body weight will be dependent on which of these terise, given the observed inconsistencies and the competing factors is more dominant. In addition, smoked marijuana effects of both increased food intake and increased energy has also been associated with fluid retention (Jones 2002), expenditure, with the additional confound of fluid retention. which may contribute to increased body weight, and mask Whilst drugs may have a similar metabolic profile, we have changes to energy balance. shown that the mechanism of effect is substance-specific. Unfortunately, no pair-feeding studies could be found that directly investigated the effect of marijuana on energy What may lead to the inconsistencies between balance. However, one study utilised pair-feeding within a epidemiology studies and animal studies? knockout model of the cannabinoid receptor type 1 (CB1), which predominantly mediates the effects of marijuana on We have presented evidence that substance abuse can affect food intake, and showed that in juvenile mice, food intake body weight, and furthermore, these changes to body weight changes solely explained body weight changes, but in adult are due to disruptions to energy balance beyond food intake mice, metabolic changes occurred independently of food alone. We acknowledge that many epidemiology studies have intake (Thornton-Jones et al. 2006). shown an inconsistent relationship between active substance The inconsistencies observed in these studies highlight abuse and body weight, which has led to the lack of scien- the potential confounds of age, species differences (which tific consensus (Nolan 2013; Sansone and Sansone 2013; may hinder extrapolation from animal models to humans), Meule 2014), but these studies can be confounded by: and dose-response relationships, when investigating the effects of substance abuse on energy balance. These studies 1. Inclusion/exclusion of alcohol - alcohol is caloric and also highlight the need to directly measure variables of can therefore directly influence body weight by being a energy balance, and not rely solely on body weight changes, source of energy intake (Mitchell and Herlong 1986). in order to avoid the potential confound of fluid retention. 2. Use of BMI - BMI (weight in kilograms/height in metres squared) as a proxy for body weight can be problematic, Grouping drugs of abuse by their metabolic profile as some drugs of abuse can simultaneously impair both height and weight, thus resulting in no net change to Collectively, these studies demonstrate that for many types BMI (Crossin et al. 2017). of drugs of abuse, there is a concurrent effect on the energy 3. Lack of repeated measures - by noting that body weight balance equation that affects body weight independently of is reduced in a substance abuse group when compared 6 R. CROSSIN ET AL.

to population controls, it cannot be determined if indi- body weight, cigarette smoking is associated with increased viduals have lost weight, or whether there has been a visceral fat and insulin resistance, which are risk factors for longer-term failure to gain weight, unless repeated meas- metabolic syndrome (Chiolero et al. 2008). Future studies ures have been taken. This is particularly relevant when will need to consider metabolic variables such as central adi- considering substance abuse in adolescence, which is a posity and insulin resistance, in addition to body weight. critical growth period (Bonjour et al. 1991). Additionally, there is a significant knowledge gap in relation 4. Age and gender – in humans the relationship between to the long-term effects of substance abuse on energy bal- substance abuse and body weight changes can be altered ance, which may persist in abstinence. As such there is a by both age and gender (Cofrancesco et al. 2007; Farhat need for longitudinal studies that continue into sustained et al. 2010; Pickering et al. 2011). Active substance abuse abstinence, in order to understand the long-term health con- is associated with weight gain in adolescent girls, but sequences of drug-induced disruptions to energy balance, not boys (Farhat et al. 2010), whereas in adults, active and the potential link to subsequent metabolic disorders, substance abuse is associated with weight loss in women including obesity and diabetes. but not men (Cofrancesco et al. 2007). Unfortunately, however, male animals predominate in the cited pre- Conclusions clinical studies, making it difficult to comment on sex- specific differences. The effects of substance abuse on body weight and the 5. Specific substance – as discussed above, the relationship causes of these changes are relatively understudied, com- between substance abuse and body weight changes are pared to other health consequences. Currently in a clinical substance-specific, therefore, understanding this rela- setting, the focus is placed on food intake as the mediator of tionship in humans is made more complex by poly-drug decreases to body weight in active substance abuse and use or by using ‘substance abuse’ as the independ- increases to body weight in abstinence, but animal models ent variable. suggest that other components of the energy balance equa- tion are contributing to these changes independently of food Meta-analysis and meta-regression may prove to be a use- intake. Disruptions to energy balance have the potential for ful tool for understanding the relationship between substance long-term health consequences for individuals, which persist abuse and body weight changes in humans, as these study even after drug use has ceased. characteristics could be incorporated. We also reinforce the importance of pair-feeding studies when using animal models to study the effects of drugs of abuse. Pair-feeding is a valuable tool for addiction biologists, The long-term health consequences of body weight to not only aid understanding of the consequences of sub- changes associated with substance abuse stance abuse on energy balance, but to control for a potential confound given that a state of positive or negative energy Despite the difficulties presented by these confounds the balance can alter the value and consumption of drugs of question of how substance abuse can affect body weight abuse (Shalev et al. 2001; Tessari et al. 2007; Jerlhag et al. remains a highly relevant one, because dramatic changes to 2009; Zheng et al. 2012). It would be easy to assume that body weight (both positive and negative) are not healthy. because many drugs of abuse reduce food intake, that the Severe and chronic weight suppression is associated with resultant changes to body weight are simply an appropriate cognitive impairments (Kretsch et al. 1997) and health physiological response to a negative energy balance. effects, including kidney and bone damage (Sharp and However it is only through pair-feeding that the true nature Freeman 1993). Furthermore, weight regain after a period of of the relationship between food intake and body weight in weight suppression is associated with increased central adi- the context of substance abuse can be revealed. posity and insulin resistance, both of which are risk factors Recognition of these effects of substance abuse on energy for the development of metabolic syndrome (Dulloo et al. balance means that treatment approaches cannot solely focus 2006). Indeed, a history of substance abuse has been identi- on food intake or nutrition. Current treatment strategies to fied as a risk factor for metabolic syndrome (Virmani et al. address weight gain in recovery from substance abuse focus 2006) potentially due to disruptions to energy balance. Thus, on nutritional education (Cowan and Devine 2012; substance abuse may contribute to the later development of Australian Drug Foundation 2016) and this will still be a obesity and metabolic disorders such as diabetes, via predis- valuable tool for restoring metabolic equilibrium (Virmani posing an individual to rapid weight regain and increased et al. 2006). However, this approach does not recognise the adiposity during abstinence. impact of energy balance disruption, which has occurred Understanding drug-induced disruptions to energy bal- during active substance abuse. Treatment strategies to ance will require studies that extend beyond the assessment address the body weight changes associated with substance of just body weight and BMI, because a ‘normal’ body abuse will need to consider the entire energy balance equa- weight is not necessarily indicative of metabolic health. tion, in order to be effective. Before appropriate treatments Individuals with a history of substance abuse may present as can be developed, further research is needed on the mecha- metabolically-obese normal-weight, a harmful phenotype nisms behind the disruption to energy balance control aris- associated with increased cardiovascular and diabetes disease ing from substance abuse, acknowledging that it is unlikely mortality (Carnethon et al. 2012). For instance, despite low that there is a single mechanism responsible for these ADDICTION RESEARCH & THEORY 7 changes to energy balance. By improving the understanding hypothalamic neuropeptide Y axis to promote weight loss. Am J – of the energy balance consequences of substance abuse, it Respir Crit Care Med. 173:1248 1254. Chiolero A, Faeh D, Paccaud F, Cornuz J. 2008. Consequences of may be possible to prevent some of the long-term adverse smoking for body weight, body fat distribution, and insulin resist- health effects, thus reducing health care costs and improving ance. Am J Clin Nutr. 87:801–809. health outcomes for individuals who have a history of sub- Cochrane C, Malcolm R, Brewerton T. 1998. The role of weight control stance abuse. as a motivation for cocaine abuse. Addict Behav. 23:201–207. Cofrancesco J, Jr Brown TT, Luo RF, John M, Stewart KJ, Dobs AS. 2007. Body composition, gender, and illicit drug use in an urban Disclosure statement cohort. Am J Drug Alcohol Abuse. 33:467–474. Collins DJ, Lapsley HM. 2008. The costs of tobacco, alcohol and illicit No potential conflict of interest was reported by the author(s). drug abuse to Australian society in 2004/05: Department of Health and Ageing Canberra. Cooper SJ, Van der Hoek GA. 1993. Cocaine: a microstructural analysis Funding of its effects on feeding and associated behaviour in the rat. Brain Res. 608:45–51. The research was supported by the Australian National Health and Cowan J, Devine C. 2008. Food, eating, and weight concerns of men in Medical Research Council (NHMRC) (940835), of which AJL is a recovery from substance addiction. Appetite. 50:33–42. Principal Research Fellow (1020737) and ZBA is a Research Fellow Cowan JA, Devine CM. 2012. Process evaluation of an environmental (1084344), the Australian Research Council (DP 110100379) of which and educational nutrition intervention in residential drug-treatment JRD was a Future Fellow during the time of the study (100100235), the facilities. Public Health Nutr. 15:1159–1167. Australian RTP scheme from which RC receives a scholarship, and the Crossin R, Cairney S, Lawrence AJ, Duncan JR. 2017. Adolescent inhal- Victorian Government’s Operational Infrastructure Support Scheme. ant abuse leads to other drug use and impaired growth; implications Funding bodies had no involvement in the design, analysis and decision for diagnosis. Aust N Z J Public Health. 41:99–104. to publish. There are no conflicts of interest or financial disclosures in Dulloo AG, Jacquet J, Seydoux J, Montani J-P. 2006. The thrifty ‘catch- this work. up fat’phenotype: its impact on insulin sensitivity during growth tra- jectories to obesity and metabolic syndrome. Int J Obes. 30:S23–S35. Ersche KD, Stochl J, Woodward JM, Fletcher PC. 2013. The skinny on ORCID cocaine: insights into eating behavior and body weight in cocaine- dependent men. Appetite. 71:75–80. Rose Crossin http://orcid.org/0000-0003-1814-1330 Farhat T, Iannotti RJ, Simons-Morton BG. 2010. Overweight, obesity, youth, and health-risk behaviors. Am J Prev Med. 38:258–267. Foltin RW, Brady JV, Fischman MW. 1986. Behavioral analysis of mari- juana effects on food intake in humans. Pharmacol Biochem Behav. References 25:577–582. Foltin RW, Fischman MW, Byrne MF. 1988. Effects of smoked mari- Audrain-McGovern J, Benowitz N. 2011. Cigarette smoking, nicotine, juana on food intake and body weight of humans living in a residen- – and body weight. Clin Pharmacol Ther. 90:164 168. tial laboratory. Appetite. 11:1–14. Australian Drug Foundation. 2016. Healthy eating during treatment for Francis H, Kraushaar N, Hunt L, Cornish J. 2011. Serotonin 5-HT4 alcohol and other drugs. In: Melbourne. receptors in the nucleus accumbens are specifically involved in the Balopole DC, Hansult CD, Dorph D. 1979. Effect of cocaine on food appetite suppressant and not locomotor effects of MDMA – intake in rats. Psychopharmacology. 64:121 122. (‘ecstasy’). Psychopharmacology. 213:355–363. Bilsky EJ, Hubbell CL, Delconte JD, Reid LD. 1991. MDMA produces a Freedman RR, Johanson C-E, Tancer ME. 2005. Thermoregulatory conditioned place preference and elicits ejaculation in male rats: a effects of 3,4-methylenedioxymethamphetamine (MDMA) in humans. modulatory role for the endogenous opioids. Pharmacol Biochem Psychopharmacology. 183:248–256. – Behav. 40:443 447. Freudenmann RW, Oxler€ F, Bernschneider -, Reif S. 2006. The origin Bonjour J-P, Theintz G, Buchs B, Slosman D, Rizzoli R. 1991. Critical of MDMA (ecstasy) revisited: the true story reconstructed from the years and stages of puberty for spinal and femoral bone mass accu- original documents . Addiction. 101:1241–1245. – mulation during adolescence . J Clin Endocrinol Metab. 73:555 563. Gambera S, Clarke J. 1976. Comments on dietary intake of drug- Brunault P, Salame E, Jaafari N, Courtois R, Reveillere C, Silvain C, dependent persons. J Am Dent Assoc. 68:155–157. Benyamina A, Blecha L, Belin D, Ballon N. 2015. Why do liver Grebenstein PE, Thompson IE, Rowland NE. 2013. The effects of transplant patients so often become obese? The addiction transfer extended intravenous nicotine administration on body weight and hypothesis. Med Hypotheses. 85:68–75. meal patterns in male Sprague-Dawley rats. Psychopharmacology. Caprioli D, Zeric T, Thorndike EB, Venniro M. 2015. Persistent palat- 228:359–366. able food preference in rats with a history of limited and extended Greenberg I, Kuehnle J, Mendelson JH, Bernstein JG. 1976. Effects of access to methamphetamine self-administration. Addict marihuana use on body weight and caloric intake in humans. Biol. 20:913–926. Psychopharmacology. 49:79–84. Carnethon MR, De Chavez PJD, Biggs ML, Lewis CE, Pankow JS, Hodgkins CC, Cahill KS, Seraphine AE, Frostpineda K, Gold MS. 2004. Bertoni AG, Golden SH, Liu K, Mukamal KJ, Campbell-Jenkins B. Adolescent drug addiction treatment and weight gain. J Addict Dis. 2012. Association of weight status with mortality in adults with inci- 23:55–65. dent diabetes. JAMA. 308:581–590. Hofstetter A, Schutz Y, Jequier E, Wahren J. 1986. Increased 24-hour Caul WF, Jones JR, Barrett RJ. 1988. Amphetamine's effects on food energy expenditure in cigarette smokers. N Engl J Med. 314:79–82. consumption and body weight: The role of adaptive processes. Behav Honma K, Honma S. 2009. The SCN-independent clocks, metham- Neurosci. 102:441. phetamine and food restriction. Eur J Neurosci. 30:1707–1717. Chen H, Hansen MJ, Jones JE, Vlahos R, Anderson GP, Morris MJ. Ignatowska-Jankowska B, Jankowski MM, Swiergiel AH. 2011. 2008. Long-term cigarette smoke exposure increases uncoupling Cannabidiol decreases body weight gain in rats: involvement of CB2 protein expression but reduces energy intake. Brain Res. receptors. Neurosci Lett. 490:82–84. 1228:81–88. Islam SN, Hossain KJ, Ahmed A, Ahsan M. 2002. Nutritional status of Chen H, Hansen MJ, Jones JE, Vlahos R, Bozinovski S, Anderson GP, drug addicts undergoing detoxification: prevalence of malnutrition Morris MJ. 2006. Cigarette smoke exposure reprograms the and influence of illicit drugs and lifestyle. BJN. 88:507–513. 8 R. CROSSIN ET AL.

Jackson T, Grilo C. 2002. Weight and eating concerns in outpatient Perkins KA. 1992. Effects of tobacco smoking on caloric intake. Br J men and women being treated for substance abuse. Eat Weight Addict. 87:193–205. Disord. 7:276–283. Petersson A, Olsson-Mortlock C, Thiblin I, Rajs J, Eksborg S. 2004. Jerlhag E, Egecioglu E, Landgren S, Salome N, Heilig M, Moechars D, Nutritional status of deceased illicit drug addicts in Stockholm, Datta R, Perrissoud D, Dickson SL, Engel JA. 2009. Requirement of Sweden—A longitudinal medicolegal study. J Forensic Sci. 49:1–10. central ghrelin signaling for alcohol reward. Proc Natl Acad Sci. Pickering RP, Goldstein RB, Hasin DS, Blanco C, Smith SM, Huang B, 106:11318–11323. Pulay AJ, Ruan WJ, Saha TD, Stinson FS. 2011. Temporal relation- Jones RT. 2002. Cardiovascular system effects of marijuana. J Clin ships between overweight and obesity and DSM-IV substance use, Pharmacol. 42:58S–63S. mood, and anxiety disorders: results from a prospective study. J Clin Kauppila T, Mecke E, Pertovaara A. 1992. Enhancement of morphine- Psychiatry. 72:1494. induced analgesia and attenuation of morphine-induced side-effects Pinter-Kubler B, Ferenczi S, Nunez C, Zelei E, Polyak A, Milanes MV, by cocaine in rats. Pharmacol Toxicol. 71:173–178. Kovacs KJ. 2013. Differential changes in expression of stress-and Keesey RE. 1989. Physiological regulation of body weight and the issue metabolic-related neuropeptides in the rat hypothalamus during of obesity. Med Clin North Am. 73:15–27. morphine dependence and withdrawal. PLoS One. 8:e67027. Keesey RE, Powley TL. 2008. Body energy homeostasis. Appetite. Piper BJ, Fraiman JB, Meyer JS. 2005. Repeated MDMA (“ecstasy”) 51:442–445. exposure in adolescent male rats alters temperature regulation, spon- Kretsch M, Green M, Fong A, Elliman N, Johnson H. 1997. taneous motor activity, attention, and serotonin transporter binding. – Cognitive effects of a long-term weight reducing diet. Int J Obes. Dev Psychobiol. 47:145 157. 21:14–21. Planeta CS, Marin M. 2002. Effect of cocaine on periadolescent rats Larue-Achagiotis C, Poussard A, Louis-Sylvestre J. 1990. Alcohol drink- with or without early maternal separation. Braz J Med Biol Res. – ing, food and fluid intakes and body weight gain in rats. Physiol 35:1367 1371. Behav. 47:545–548. Rahminiwati M, Nishimura M. 1999. Effects of. DELTA. 9- Lenoir M, Serre F, Cantin L, Ahmed SH. 2007. Intense sweetness sur- Tetrahydrocannabinol and Diazepam on Feeding Behavior in Mice. J – passes cocaine reward. PLoS One. 2:e698. Vet Med Sci. 61:351 355. LeSage MG. 2009. Toward a nonhuman model of contingency manage- Rodondi N, Pletcher MJ, Liu K, Hulley SB, Sidney S. 2006. Marijuana ment: effects of reinforcing abstinence from nicotine self-administra- use, diet, body mass index, and cardiovascular risk factors (from the – tion in rats with an alternative nondrug reinforcer. CARDIA study). Am J Cardiol. 98:478 484. Psychopharmacology. 203:13–22. Sansone RA, Sansone LA. 2013. Obesity and substance misuse: is there ‘ ’ a relationship? Innov Clin Neurosci. 10:30–35. Linnemann T, Wall T. 2013. This is your face on meth : The punitive ‘ ’ Santolaria-Fernandez FJ, Gomez-Sirvent J, Gonzalez-Reimers CE, spectacle of white trash in the rural war on drugs. Theor Criminol. ı ı – Batista-Lopez J, Jorge-Hernandez J, Rodr guez-Moreno F, Mart nez- 17:315 334. Lupien JR, Bray GA. 1988. Nicotine increases thermogenesis in brown Riera A, Hernandez-Garcia MT. 1995. Nutritional assessment of drug addicts. Drug Alcohol Depend. 38:11–18. adipose tissue in rats. Pharmacol Biochem Behav. 29:33–37. Santolaria F, Perez-Manzano JL, Milena A, Gonzalez-Reimers E, Luz J, Griggio M, Plapler H, De-Meo-Bancher M, Carvalho-Kosmiskas J. Gomez-Rodr {guez MA, Mart{nez-Riera A, Aleman-Valls MR, de la 1996. Effects of ethanol on energy balance of rats and the inappropri- Vega-Prieto MJ, 2000. Nutritional assessment in alcoholic patients. ateness of intraperitoneal injection. Alcohol. 13:575–580. Its relationship with alcoholic intake, feeding habits, organic compli- Makisumi T, Yoshida K-i, Watanabe T, Tan N, Murakami N, cations and social problems. Drug Alcohol Depend. 59:295–304. Morimoto A. 1998. Sympatho-adrenal involvement in methampheta- Shalev U, Yap J, Shaham Y. 2001. Leptin attenuates acute food depriv- mine-induced hyperthermia through skeletal muscle hypermetabo- ation-induced relapse to heroin seeking. J Neurosci. 21:1–5. lism. Eur J Pharmacol. 363:107–112. Sharp C, Freeman C. 1993. The medical complications of anorexia Meule A. 2014. The relation between body mass index and substance nervosa. Br J Psychiatry. 162:452–462. use: a true can of worms. Innovat Clin Neurosci. 11:11. Singh RA, Kosten TA, Kinsey BM, Shen X, Lopez AY, Kosten TR, Mitchell MC, Herlong HF. 1986. Alcohol and nutrition: caloric value, Orson FM. 2012. Dose-dependent changes in the locomotor bioenergetics, and relationship to liver damage. Annu Rev Nutr. – responses to methamphetamine in BALB/c mice: Low doses induce 6:457 474. hypolocomotion. Pharmacol Biochem Behav. 103:230–236. Mohs ME, Watson RR, Leonard-Green T. 1990. Nutritional effects of Sumithran P, Proietto J. 2013. The defence of body weight: a physiological marijuana, heroin, cocaine, and nicotine. J Am Dent Assoc. basis for weight regain after weight loss. Clin Sci. 124:231–241. – 90:1261 1267. Tessari M, Catalano A, Pellitteri M, Di Francesco C, Marini F, Gerrard Mysels DJ, Vosburg S, Benga I, Levin FR, Sullivan MA. 2011. Course of PA, Heidbreder CA, Melotto S. 2007. Preclinical Study: Correlation weight change during naltrexone vs. methadone maintenance for between serum ghrelin levels and cocaine-seeking behaviour trig- opioid-dependent patients. J Opioid Manag. 7:47. gered by cocaine-associated conditioned stimuli in rats. Addict Biol. Naghavi M, Wang H, Lozano R, Davis A, Liang X, Zhou M, Vollset 12:22–29. SEV, Abbasoglu Ozgoren A, Norman RE, Vos T. 2015. Global, Thornhill J, Hirst M, Gowdey C. 1976. Disruption of diurnal feeding – regional, and national age sex specifi c all-cause and cause-specifi patterns of rats by heroin. Pharmacology Biochemistry and Behavior. – c mortality for 240 causes of death, 1990 2013: a systematic ana- 4:129–135. lysis for the Global Burden of Disease Study 2013. Lancet. Thornton-Jones ZD, Kennett GA, Benwell KR, Revell DF, Misra A, 385:117–171. Sellwood DM, Vickers SP, Clifton PG. 2006. The cannabinoid CB 1 Natividad LA, Torres OV, Friedman TC, O’Dell LE. 2013. receptor inverse agonist, rimonabant, modifies body weight and adi- Adolescence is a period of development characterized by short-and ponectin function in diet-induced obese rats as a consequence of long-term vulnerability to the rewarding effects of nicotine and reduced food intake. Pharmacol Biochem Behav. 84:353–359. reduced sensitivity to the anorectic effects of this drug. Behav United Nations Office of Drugs and Crime. 2014. World Drug Report. Brain Res. 257:275–285. New York. (Sales No. E.14.XI.7). Neale J, Nettleton S, Pickering L, Fischer J. 2012. Eating patterns Vetter CS, Doremus -Fitzwater TL, Spear LP. 2007. Time course of ele- among heroin users: a qualitative study with implications for nutri- vated ethanol intake in adolescent relative to adult rats under con- tional interventions. Addiction. 107:635–641. tinuous, voluntary-access conditions. Alcoholism Clin Exp Res. Nolan LJ. 2013. Shared urges? The links between drugs of abuse, eating, 31:1159–1168. and body weight. Curr Obes Rep. 2:150–156. Virmani A, Binienda Z, Ali S, Gaetani F. 2006. Links between nutrition, Nolan LJ, Stolze MR. 2012. Drug use is associated with elevated food drug abuse, and the metabolic syndrome. Ann N Y Acad Sci. consumption in college students. Appetite. 58:898–906. 1074:303–314. ADDICTION RESEARCH & THEORY 9

Warren CS, Lindsay AR, White EK, Claudat K, Velasquez SC. 2013. Williams CM, Rogers PJ, Kirkham TC. 1998. Hyperphagia in pre-fed Weight-related concerns related to drug use for women in substance rats following oral D 9-THC. Physiol Behav. 65:343–346. abuse treatment: Prevalence and relationships with eating pathology. Williams MT, Moran MS, Vorhees CV. 2004. Behavioral and J Subst Abuse Treat. 44:494–501. growth effects induced by low dose methamphetamine administra- World Health Organisation. 2014. Global status report on noncommu- tion during the neonatal period in rats. Int J Dev Neurosci. nicable diseases 2014. 22:273–283. Wiley JL, Burston JJ, Leggett DC, Alekseeva OO, Razdan RK, Zheng D, de Vaca SC, Carr KD. 2012. Food restriction increases acqui- Mahadevan A, Martin BR. 2005. CB1 cannabinoid receptor-medi- sition, persistence and drug prime-induced expression of a cocaine- ated modulation of food intake in mice. Br J Pharmacol. conditioned place preference in rats. Pharmacol Biochem Behav. 145:293–300. 100:538–544. Williams CL, Bihm CC, Rosenfeld GC, Burks TF. 1997. Morphine tol- Zwillich CW, Doekel R, Hammill S, Weil JV. 1978. The Effects of erance and dependence in the rat intestine in vivo. J Pharmacol Exp Smoked Marijuana on Metabolism and Respiratory Control 1–3. Am Ther. 280:656–663. Rev Respir Dis. 118:885–891. Rose Crossin (737900)

Appendix 2 – A human cohort of adolescent inhalant users

Introduction This chapter describes the design for a human cohort of inhalant users, which has been designed, has ethics approval, and is in the early stages of patient recruitment. This study will build upon findings from Chapters 2 and 3, in which a significant association was identified between adolescent inhalant use and impaired height and weight, and Chapter 4, in which it was found that growth was impaired in an animal model of adolescent inhalant abuse, and also showed that inhalant-induced appetite suppression is associated with altered levels of appetite-regulating hormones, but that reduced food intake could not fully account for the observed growth impairments. This study provides the opportunity to translate these findings back into a clinical population, to assess the relationship between inhalant abuse, food intake, growth, and relevant hormone levels. This study will be the first to investigate growth and food intake, including potential mechanisms, in a clinical population of adolescents who misuse inhalants. This study may lead to improved treatment strategies for adolescents who misuse inhalants and provide improved information to adolescents on the possible health risks of inhalant misuse, thus improving prevention. The aims of this study are to characterise the effects of adolescent inhalant abuse on growth, body composition, and food intake, and to identify whether effects on food intake are related to alterations in the circulating levels of appetite regulating hormones and glucose regulation. Thus, this study seeks to determine the validity of the findings from Chapter 4, in a clinical setting.

Methods This study is being conducted in collaboration with the Adolescent Drug and Alcohol Withdrawal Service (ADAWS), which is part of Mater Health in Brisbane, Australia. The rationale for the study location was that ADAWS have a relatively high number of clients (approximately 24 per year) for whom their primary substance of concern was inhalants, and it allowed for targeted recruitment of adolescents and young adults. Furthermore, it allowed for a multiple control group design, whereby inhalant users could be compared to both age and gender-matched other drug users (to identify effects specific to inhalants) and non-drug using adolescents (recruited from the community). Lastly, ADAWS conducts a residential service with clients entering into voluntary treatment for 14 days, and upon entering the service, an incoming health check is conducted, including blood tests for blood-borne diseases. Therefore, data collection for this study could be appended to the incoming health check with minimal disruption to both staff and clients.

This research will be conducted as an analytical observational study, with a cross-sectional cohort. The study will have three groups. The primary study group will be adolescents (aged 12-20) with inhalants identified as their primary substance of concern. A secondary study group will be adolescents where the primary substance of concern is not inhalants (e.g. marijuana, alcohol, or methamphetamines). This group will enable differentiation between the effects of substance use in adolescence, from the specific effects of inhalants. The control group will be made up of adolescents with no history of substance use. As participants will be allocated into groups based on their substance use history, the study is not able to be randomised.

157 Rose Crossin (737900)

This study design has been selected in order to differentiate between substance use in adolescence and the specific effects of inhalants. Each participant will have one observation made, where data on drug use history, growth and food intake will be collected and a blood sample will be taken for the hormone studies. The primary data collection process will be cross-sectional, which was chosen in order to collect data concurrently with participants seeking treatment for their substance use. The research question is intended to determine the effects of active / current substance use, rather than the long-term effects or effects in abstinence, thus there is no intention to collect longitudinal data. However, in order to assess current growth against previous growth trajectory, historical data will be sought from childhood heath records. Depending on the outcomes of this study, a further longitudinal study may be developed in order to consider the long-term effects on growth and metabolism arising from adolescent inhalant abuse.

The primary study population will be clients of ADAWS, aged from 12-20, of both sexes. Inhalant abuse predominantly occurs in adolescence and in both sexes; therefore, the decision was made to exclude participants aged over 20 but to include both males and females. In addition, the study is intended to investigate the effects of inhalants during a critical growth period (the adolescent growth spurt), thus the age group has been selected in order to encompass this peak growth phase. The primary study group will be adolescents for whom inhalants in their main substance use of concern, which will be determined by self-reporting and supported by ADAWS staff assessment. All types of inhalants (e.g. glue, petrol, or solvents) will be eligible for inclusion, so that results are applicable to inhalant use broadly, rather than a single type of inhalant. Clients who meet these eligibility criteria will be invited to participate in the study, which is voluntary. The secondary study group will be adolescents for whom a substance other than inhalants is their primary concern, determined by self-reporting and supported by ADAWS staff assessment. Control participants will be adolescents with no history of substance use (including alcohol and cigarettes). As inhalant use is associated with vulnerable youth and low socio-economic status, control participants will be sought from a similar geographic area to the primary study group. Approximate age and gender matching will be undertaken, where possible within the constraints of volunteer availability. Church groups and youth groups will be approached to invite individuals to act as control participants, and participants may also nominate friends or siblings to act as control participants. An Expression of Interest document will be sent to groups, with a request that interested individuals contact the study coordinator. Interested individuals will then be provided with an Information Sheet and Consent Form and will have the risks and benefits verbally explained to them. Control participants will also require parent / guardian consent if they are under 18 years of age.

Participation will be voluntary and require informed consent, including parent/guardian consent for participants aged under 18 years. Exclusion criteria include being unable to provide informed consent, participation considered harmful by the ADAWS team, or non-adherence. Furthermore, individuals who have been previously (i.e. prior to drug misuse) diagnosed with type 1 or type 2 diabetes will be excluded from the study. This is due to the fact that the study will measure levels of appetite regulating hormones such as insulin, and a history of diabetes and / or for managing diabetes will confound the study results. Pregnant women will be excluded from the study as pregnancy can alter metrics of body composition, growth, food intake, and hormone levels, which would confound the results of this study.

158 Rose Crossin (737900)

The primary outcomes are:

1. Height (measured in cm) 2. Body weight (measured in kg)

Secondary outcomes are:

1. Demographic data (measured by a questionnaire) 2. Drug use history (measured by a questionnaire) 3. Food intake frequency (as a proxy for total dietary intake) (measured by a questionnaire) 4. Food preferences (measured by a questionnaire) 5. Adiposity (measured by waist circumference and skinfold tests) 6. Feelings of hunger (measured by questionnaire) 7. Levels of circulating appetite regulating hormones (measured from blood samples) 8. Glucose regulation (measured by a glucose tolerance test (GTT))

Four instruments will be used for data collection:

1. A questionnaire that has been designed for this study, to collect information on drug use history and food intake 2. Direct measurement of body growth and composition (height, weight, waist circumference, skin fold tests for adiposity), which will be entered by an ADAWS staff member on to the questionnaire 3. Review of childhood patient health records (if available) to determine a participant’s historic growth trajectory 4. Blood specimen collection, which includes the GTT

It is important for this study that a detailed drug use history is taken. Given the finding that height impairments associated with adolescent inhalant abuse are affected by the duration of inhalant use (Chapter 2), the duration of substance abuse is pertinent. Additionally, as the study is investigating substance abuse that is occurring during the adolescent growth spurt, it will be important to determine when substance use began. Lastly, different substances of abuse can have markedly different effects on body weight and food intake, thus it is important to collect data on each type of substance separately. Food and drink intake data will be collected by a questionnaire, where participants are asked to reflect on food and drink intake from the prior two days. Whilst best practice would be a detailed food diary, this was deemed to be unfeasible for this study and likely to yield unreliable results, given the age and substance use concerns of these participants. Instead, a food and drink frequency questionnaire will be utilised, to investigate how often different types of food are eaten, dietary composition, whether caloric intake is dominated by food or drink, the typical meal size, and food preferences. These are all important components as inhalant use has been shown to not only affect food intake, but also food preferences. A question regarding feelings of hunger when under the influence of substances of concern is included in order to determine if hunger is acutely affected by substance use. Lastly, a question relating to feelings of hunger is included, in order to provide a potential behavioural link to the levels of appetite-regulating hormones from the blood tests. Examples for meal sizes and meal types have been provided in

159 Rose Crossin (737900) the questionnaire, to reduce inter-participant variability in interpretation of the categories and thus improve data quality.

Growth and body composition will be assessed by direct measurement of height, weight, waist circumference, and skin fold tests for adiposity. These are all simple and standard growth measures, which will allow comparison to population growth percentiles. BMI will be calculated from height and weight data. Whether an individual meets Failure to Thrive (FTT) criteria will be determined by comparison of height and weight data to population growth percentiles. Standard FTT criteria will be used; weight below the 3rd percentile or 20% below ideal weight for height. Blood tests including the GTT will be undertaken by a trained nurse, in conjunction with other standard blood tests used as part of the ADAWS in-patient health check. Appetite-regulating hormone levels are standard tests and are able to be analysed by Mater Pathology, in accordance with standard protocols. All data will be collected by trained ADAWS staff members, who are experienced in collecting drug use history and general health data. A specific instruction sheet on taking skin fold measurements for adiposity will be provided. Given the simplicity of the growth measures and the standardised questionnaire, there is no intention to assess inter-rater variability.

Based on ADAWS historical data, approximately 24 people per year are admitted due to a primary inhalant use issue. Assuming a conservative 50% recruitment rate, over 18 months, we estimate that the primary study group will have approximately 20 participants. Thus, the total number of participants will be approximately 60 across the three groups. The appropriateness of this group size was determined using power calculations based on data from Chapter 2. The effect size was calculated as 0.38, based upon the height and weight data. Assuming a Repeated Measure ANOVA for height and weight (two variables), with a standard type 1 error of 0.05 and a conservative type 2 error of 0.90, across three groups, the required total sample size is 69. Thus, this matches with our estimated sample size of approximately 60 participants (20 per group).

The full and approved research protocol for this study is provided as Appendix 3 to this thesis and the approved questionnaire is provided as Appendix 4.

Current status The project was approved by the Mater Human Research Ethics Committee in July 2016 HREC/16/MHS/32. The Shared Services Agreement to conduct research in the clinical setting was approved on 27 January 2017 (RG-16-279). Recruitment into the study has been slower than expected, in part due to a lack of suitable clients for recruitment, and also due to a lower than expected recruitment rate when suitable clients have been identified. The study will continue throughout 2018, but at the time of writing, only 4 inhalant using clients have participated in the study (Group 1 in the study). These results are not reported in the thesis.

Preliminary findings As yet, there is insufficient data for conclusions to be reached, or for statistical testing to be undertaken. However, it is interesting to note that so far, all participants have a body weight less than the 10th percentile of weight-for-age on standard growth charts, and a BMI less than the 5th percentile of BMI-for-age, relative to their gender. This would suggest that growth

160 Rose Crossin (737900) findings are consistent with results from Chapters 2-4. Furthermore, participants have reported low food intakes, with frequent skipped meals and small meal sizes, suggesting that inhalant abuse is resulting in a reduction in food intake, consistent with Chapter 4. Lastly, all participants to date report that after using inhalants that they do not have feelings of hunger, suggesting that inhalants have a physiological appetite-suppressing effect. This study will be published when completed, and will provide an understanding of whether the previous findings (Chapter 4) are clinically valid.

161 Rose Crossin (737900)

Appendix 3 – Research Protocol for the human cohort study The approved research protocol is appended for additional information to the human cohort study described in Appendix 2.

162

Table of contents 1. INTRODUCTION ...... 5 2. BACKGROUND ...... 5 3. AIMS OF STUDY ...... 8 4. OBJECTIVES ...... 8 4.1 Primary Objectives ...... 8 4.2 Secondary Objective ...... 8 5. HYPOTHESES ...... 8 5.1 Primary Hypotheses ...... 8 5.2 Secondary Hypothesis ...... 8 6. STUDY DESIGN ...... 9 7. STUDY SETTING/LOCATION(S) ...... 9 8. STUDY DURATION ...... 9 9. STUDY POPULATION ...... 9 9.1 Inclusion criteria ...... 10 9.2 Exclusion criteria ...... 10 9.3 Potential for risk, burdens and benefits to participants ...... 10 10. STUDY OUTCOMES ...... 11 10.1 Primary Outcomes ...... 11 10.2 Secondary Outcomes ...... 11 11. STUDY PROCEDURES ...... 11 11.1 Recruitment and consent of participants ...... 11 11.2 Withdrawal of participants from a study ...... 12 11.3 Randomisation ...... 12 11.4 Measurement tools used ...... 12 11.5 Study involvement by participants ...... 14 11.6 Data management and storage ...... 14 11.7 Safety considerations/Patient safety ...... 15 11.8 Data monitoring ...... 15 12. SAMPLE SIZE AND DATA ANALYSIS ...... 15 12.1 Sample size and statistical power ...... 15 12.2 Data analysis plan ...... 16 13. ETHICAL CONSIDERATIONS ...... 16 14. DISSEMINATION OF RESULTS AND PUBLICATIONS ...... 16

Page 1 of 20

15. OUTCOMES AND SIGNIFICANCE ...... 17 16. BUDGET ...... 17 17. GLOSSARY OF ABBREVIATIONS ...... 18 18. REFERENCES ...... 18

Page 2 of 20

FULL STUDY TITLE The effects of adolescent inhalant abuse on growth and food intake

SHORT TITLE Inhalants and growth

LAY DESCRIPTION OF THE PROJECT Inhalant abuse is when people intentionally inhale substances containing toluene (e.g. petrol, thinners, or glue) in order to get ‘high’. Young people (aged 12–20) are the main users of inhalants, with 20% of 12-13 year olds in Australia saying they have abused inhalants at least once. Common side effects of inhalant misuse are reduced body weight and suppressed appetite; however the processes leading to these outcomes remain unknown. However, these side effects have the potential to cause both immediate and long-term health risks, particularly as adolescence is a critical growth period. There is also the potential for inhalant misuse to continue to impact food preferences and growth, even after use has ceased. This research project will investigate growth, food intake, and appetite regulating hormones, in a human population of adolescents who are seeking treatment for inhalant abuse.

RELEVANT LEGISLATION AND GUIDANCE This study will be conducted to ensure that at all times it complies with:  Catholic Health Australia (2001). Code of Ethical Standards for Catholic Health and Aged Care Services in Australia;  Current best practice in ethics including abiding by the National Statement and all other relevant NHMRC standards;  Relevant State and Commonwealth Acts and legislations;  Relevant Institutional policies and procedures (available on Mater DocuCube).

STUDY INVESTIGATORS Name Phone Email Institution Study Role (e.g. Principal Investigator) Dr Jhodie 03 9035 [email protected] Florey Institute of Principal investigator Duncan 6731 Neuroscience and Mental Health Ms Nicole 07 3163 [email protected] Mater Health Coordinating local

Inhalants and growth study protocol Dr Jhodie Duncan Protocol Version 1.1 Date: 21/07/2016 Page 3 of 20

Lacey 8400 Service ADAWS investigator Ms Amanda 07 3163 [email protected] Mater Health Associate Tilse 8400 Service ADAWS Investigator Mrs Rose 03 9035 [email protected] Florey Institute of Coordinating Crossin 6669 Neuroscience and Investigator / PhD Mental Health student A/Prof Leanne 07 3138 [email protected] Queensland Associate Hides 6144 University of Investigator Technology

Page 4 of 20 Inhalants and growth Dr Jhodie Duncan < AU/1/1F86217> < MR-2016-89> Protocol Version 1.1 Date: 21/07/2016

1. INTRODUCTION Inhalant abuse is the intentional inhalation of products containing the volatile solvent toluene (e.g. petrol, thinners or glue). Inhalant abuse is prevalent in adolescent populations, particularly in vulnerable youth. Low body weight and appetite suppression are commonly reported side effects of inhalant abuse, though the processes leading to these outcomes remain unknown. Furthermore, it is not known whether the observed changes in growth are solely attributable to reduced food intake, or whether there are concurrent changes to metabolism in affected individuals. These consequences of adolescent inhalant abuse have the potential to cause significant harm, as adolescence is a critical growth period and any disruption to growth can persist into adulthood. Furthermore, altered eating behaviour during adolescence, particularly changes to dietary preferences, may contribute to subsequent health concerns such as metabolic syndrome. Members of the study team have conducted a retrospective analysis from a cohort of males who used inhalants during adolescence and found a significant association between inhalant use and impaired height and weight. This was then further verified using an animal model of adolescent inhalant abuse, which confirmed that growth was impaired, and also showed that inhalant-induced appetite suppression is associated with altered levels of appetite-regulating hormones, but that reduced food intake could not fully account for the observed growth impairments. This study provides the opportunity to translate these findings back into a human population, to assess the relationship between inhalants, food intake, growth, and hormone levels. This study will be the first to investigate growth and food intake, including potential mechanisms, in a human population of adolescents who misuse inhalants. Data will be collected by a self-reported questionnaire on drug use history and food intake, direct measures of growth and body composition, and measurement of appetite-regulating hormones from blood samples. This study may lead to improved treatment strategies for adolescents who misuse inhalants and provide improved information to adolescents on the possible health risks of inhalant misuse, thus improving prevention.

2. BACKGROUND Inhalant misuse is a form of substance abuse and involves the intentional inhalation of vapours from household or industrial products, in order to create a feeling of euphoria and an altered mental state [1]. Products that are commonly abused as inhalants include petrol, spray paint, thinners, and glue. These products are cheap, legal to purchase, and readily accessible, which is thought to add to their attractiveness as substances of abuse [2]. One commonality of these products is that they all contain the primary volatile solvent toluene. The reinforcing nature of toluene [3, 4] is thought to underlie the addictive drive to continue abusing inhalants. Thus, while inhaled products are not technically considered ‘drugs’, they share the reinforcing nature of many drugs of abuse.

Historically in Australia inhalant abuse, especially petrol sniffing, has been associated with Indigenous communities. Inhalant abuse still remains a significant public health issue for Indigenous Australians [5] with abuse rates of up to 60% in some remote communities [6]. In recent years though, inhalant abuse has become more prevalent among young adolescent populations. Inhalants are one of the first and most common substance abused by the 12-13 age

Inhalants and growth study protocol Dr Jhodie Duncan Protocol Version 1.1 Date: 21/07/2016 Page 5 of 20 group in Australia [7] with 20% of 12 year-olds having experimented with inhalants [8]. Furthermore, this pattern of early adolescent use is reflected globally [9]. Inhalant abuse is also associated with low socioeconomic status [10] and inhalant abusers are disproportionately represented in the mental health, juvenile justice, and protective services systems [2]. Therefore, given that much of the use data on inhalant abuse comes from secondary school surveys, it is likely that inhalant abuse rates are actually under-reported in Australia. Of further concern, a recent survey by the Australian government found that inhalant abuse in Australia is increasing, with a 23% overall increase in use between 2007-2011 compared to a 4% reduction in alcohol and smoking [7] over the same period, highlighting the growing popularity of this form of substance abuse.

Adolescence is defined as the period between puberty and adulthood, typically between the ages of 12 and 20 years of age [11]. During this period, critical maturational processes occur including a range of physiological, hormonal and psychological processes. Exposure to harmful substances during adolescence can disrupt these processes and has the potential for long-term health consequences [12, 13]. For example, adolescence encompasses an intensive period of growth known as the adolescent growth spurt [14] where skeletal structures grow and mature [15]. Indeed, in humans, over half of the peak bone mass is attained during the adolescent growth spurt [15, 16]. However, the adolescent growth spurt can be disrupted by factors such as malnutrition [17]. Growth impairments that arise in childhood have the potential to recover if the harmful factor is removed, but this potential is reduced as age increases, such that adolescents usually only achieve partial recovery and no recovery is possible into adulthood [18]. Thus, any disruption to growth during adolescence has the potential to cause long-term effects on growth patterns [19]. Given that the highest rates of inhalant abuse are occurring during a critical maturational period, it highlights the importance of studying the impacts of inhalant abuse specifically in adolescents.

One of the most profound, but under-explored, consequences of inhalant abuse is its effect on food intake, body weight, and growth; especially if exposure occurs during adolescence, suggesting inhalants result in metabolic dysfunction. In humans, adolescent inhalant abuse is associated with disordered eating [20], weight loss [21], and emaciation [22]. Furthermore, unpublished data (manuscript in review) from study investigators has shown that adolescent inhalant abuse is associated with meeting Failure to Thrive (FTT) criteria, because of the severity of the weight impairment. These findings are consistent with rodent studies, with adolescent toluene exposure associated with reduced food intake, decreased weight gain, and reduced adiposity [23-25]. Although toluene is often associated with decreased food intake, toluene exposure during adulthood in a workplace setting is also associated with altered dietary preferences, including an increased preference for carbohydrates [26]. Additionally, a preference for sweet and fatty foods has been observed in adolescent toluene-exposed rats and this change persists into abstinence [27].

In humans there is evidence that incidental inhalation of petrol fumes may have an acute appetite suppressing effect, and the study’s authors hypothesised that in a food-constrained environment, a desire to suppress feelings of hunger could be a driver for intentional petrol sniffing behaviour [28]. This data confirmed previous anecdotal reports that petrol sniffing in Australian Indigenous communities is partly driven by a desire to suppress appetite [29]. In contrast, a

Page 6 of 20 Inhalants and growth Dr Jhodie Duncan < AU/1/1F86217> < MR-2016-89> Protocol Version 1.1 Date: 21/07/2016 human study of workplace exposure to toluene found that chronic exposure was associated with an increased food intake, though this was not reflected in an increased body mass index (BMI) [26]. The authors hypothesised that toluene exposure resulted in a higher basal metabolic rate, though this was not tested [26] . In adult rats, toluene- induced anorexia has been attributed in part to a decrease in the hypothalamic expression of Neuropeptide Y (NPY), a peptide which stimulates food intake, which was associated with the observed decrease in food intake in this study [23]. Whilst human data regarding food intake is limited, these studies suggest that inhalant abuse may affect appetite and food intake; however the underlying mechanisms of these effects are unknown.

There is evidence to suggest that inhalant abuse can impair metabolic processes, because the normal responses to disrupted energy homeostasis do not occur. Exposure to toluene during adolescence impairs weight gain and this effect persists into abstinence, however, food intake is reduced even further than what can be attributed to reduced body weight [27]. This finding suggests that there is impairment to the metabolic feedback processes, which should act to restore homeostasis in response to insufficient energy intake. In rats, adolescent exposure to toluene has been shown to alter levels of gut hormones, including insulin, amylin, and Peptide YY (PYY), as well as decrease hypothalamic leptin receptor mRNA expression [27], suggesting that appetite suppression may be due to impaired hormonal signalling.

In addition to effects on food intake and food preferences, toluene is associated with impaired growth. In weanling rats, chronic toluene exposure has been shown to impair skeletal growth, with a significant reduction in rump width and variable reduction in torso length [30]. Skeletal variations have also been shown in the offspring of rats where there was maternal exposure to toluene, as well as decreased body weight in the offspring [31]. In humans, regular inhalant abuse during pregnancy is also associated with neonatal skeletal malformations and growth impairment [32]. In addition, unpublished data (manuscript in preparation) from study investigators shows that adolescent inhalant abuse is associated with impaired height, which persisted into adulthood, even after inhalant abuse ceased.

Some of the metabolic effects arising from adolescent inhalant abuse, including reduced body weight, persist into abstinence [27]. Additionally, the preferential consumption of a high fat, high sugar diet perpetuates the metabolic dysfunction caused by adolescent inhalant abuse and prevents metabolic normalisation in abstinence [27]. These changes, despite the fact that exposure has ceased, have important clinical significance as they may increase the risk of adult-onset disorders. For example in humans, a preference for unhealthy foods is known to increase the risk for metabolic disorders such as type 2 diabetes [33, 34]. Chronic exposure to low levels of toluene in a workplace setting has also been associated with insulin resistance [35]. Insulin resistance is the hallmark of Metabolic Syndrome: a group of abnormalities associated with the development of type 2 diabetes [36]. Adolescent inhalant abuse could therefore be initiating a sequence of altered food intake, long-term metabolic dysfunction, and lead to a chronic disease burden for individuals.

Page 7 of 20 Inhalants and growth Dr Jhodie Duncan < AU/1/1F86217> < MR-2016-89> Protocol Version 1.1 Date: 21/07/2016

Collectively, these studies suggest that toluene exposure can lead to symptoms of metabolic dysfunction (e.g. appetite suppression and changes to appetite-regulating hormones) and impaired growth. However, both the mechanisms underlying the metabolic and growth impacts of adolescent inhalant abuse and the long-term health effects of these changes are unknown. Thus this project will address an important knowledge gap about the metabolic and growth impacts of adolescent inhalant abuse. Ultimately it is hoped that this information will assist clinical practitioners to identify and manage high-risk individuals with a history of inhalant abuse, subsequently improving their quality of life.

3. AIMS OF STUDY The aims of this study are to characterise the effects of adolescent inhalant abuse on growth and food intake (as both a metabolic variable and driver of growth), and to identify whether effects on food intake are related to alterations in the circulating levels of appetite regulating hormones and glucose regulation.

4. OBJECTIVES

4.1 Primary Objectives 1. To determine the effects of adolescent inhalant abuse on growth by measuring metrics of growth and body composition, including height, weight, adiposity, and waist circumference, and comparing current growth to childhood growth trajectories, control group, and population growth charts. 2. To determine the effects of adolescent inhalant abuse on food intake by collecting data on food consumption and dietary preferences and comparing this to a control group.

4.2 Secondary Objective 1. To determine whether adolescent inhalant abuse is associated with altered levels of appetite regulating hormones (e.g. leptin, insulin, and ghrelin) by measuring circulating levels of these hormones. 2. 2 To determine whether adolescent inhalant abuse is associated with altered glucose regulation by undertaking a glucose tolerance test (GTT).

5. HYPOTHESES

5.1 Primary Hypotheses We hypothesise that adolescent inhalant abuse will impair growth, resulting in reduced weight and height compared to both adolescents not misusing substances of abuse (controls) and adolescents who use substances other than inhalants. We further hypothesise that adolescent inhalant abuse will result in a reduced food intake and altered dietary preferences.

5.2 Secondary Hypothesis We hypothesise that inhalant-induced appetite suppression will be associated with altered levels of appetite regulating hormones, such as leptin, insulin, and ghrelin, in addition to altered glucose regulation.

Page 8 of 20 Inhalants and growth Dr Jhodie Duncan < AU/1/1F86217> < MR-2016-89> Protocol Version 1.1 Date: 21/07/2016

6. STUDY DESIGN This research will be conducted as an analytical observational study, with a cross-sectional cohort. The study will have three groups. The primary study group will be adolescents (aged 12-20) with inhalants identified as their primary substance of concern. A secondary study group will be adolescents where the primary substance of concern is not inhalants (e.g. marijuana, alcohol, or methamphetamines). This group will enable differentiation between the effects of substance use in adolescence, from the specific effects of inhalants. The control group will be made up of adolescents with no history of substance use. As participants will be allocated into groups based on their substance use history, the study is not able to be randomised.

This study design has been selected in order to differentiate between substance use in adolescence and the specific effects of inhalants. Each participant will have one observation made (i.e. this is not a longitudinal study), where data on drug use history, growth and food intake will be collected and a blood sample will be taken for the hormone studies. The primary data collection process will be cross-sectional, which was chosen in order to collect data concurrently with participants seeking treatment for their substance use. The research question is intended to determine the effects of active / current substance use, rather than the long-term effects or effects in abstinence, thus there is no intention to collect longitudinal data. However, in order to assess current growth against previous growth trajectory, historical data will be sought from childhood heath records. Depending on the outcomes of this study, a further longitudinal study may be developed in order to consider the long-term effects on growth and metabolism arising from adolescent inhalant abuse.

7. STUDY SETTING/LOCATION(S) This study will be conducted at the Mater Health Service Adolescent Drug and Alcohol Withdrawal Service (ADAWS), with administration and data analysis also being conducted by the Florey Institute of Neuroscience and Mental Health. The Queensland University of Technology will provide assistance with interpretation of results. Primary local coordination will be provided by ADAWS, with overall coordination being undertaken by the Florey Institute. The study has been designed to fit in with the current health check that is conducted when clients of ADAWS are admitted, in order to reduce the burden on ADAWS staff.

8. STUDY DURATION Data collection will be taken opportunistically, when ADAWS clients who meet the inclusion criteria are admitted. It is estimated that in order to meet the desired number of participants, the study will run for 18 months (from June 2016 to December 2017). The data collection process (body composition and questionnaire) for each participant will take less than 1 hour, with a 2 hour period required for the GTT. Data analysis will be conducted at the end of the data collection period, and we estimate that the outcomes from this study will be published in early 2018.

9. STUDY POPULATION The primary study population will be clients of ADAWS, aged from 12-20, of both sexes. Inhalant abuse predominantly occurs in adolescence and in both sexes; therefore, the decision was made to exclude participants aged over 20 but to

Page 9 of 20 Inhalants and growth Dr Jhodie Duncan < AU/1/1F86217> < MR-2016-89> Protocol Version 1.1 Date: 21/07/2016 include both males and females. In addition, the study is intended to investigate the effects of inhalants during a critical growth period (the adolescent growth spurt), thus the age group has been selected in order to encompass this peak growth phase.

The primary study group will be adolescents for whom inhalants in their main substance use of concern, which will be determined by self-reporting and supported by ADAWS staff assessment. All types of inhalants (e.g. glue, petrol, or solvents) will be eligible for inclusion, so that results are applicable to inhalant use broadly, rather than a single type of inhalant. Clients who meet these eligibility criteria will be invited to participate in the study, which is voluntary.

The secondary study group will be adolescents for whom a substance other than inhalants is their primary concern, determined by self-reporting and supported by ADAWS staff assessment.

Control participants will be adolescents with no history of substance use (including alcohol and cigarettes). As inhalant use is associated with vulnerable youth and low socio-economic status, control participants will be sought from a similar geographic area to the primary study group. Approximate age and gender matching will be undertaken, where possible within the constraints of volunteer availability. Church groups and youth groups will be approached to invite individuals to act as control participants, and participants may also nominate friends or siblings to act as control participants.

9.1 Inclusion criteria Inclusion criteria are adolescents aged 12-20 who are ADAWS clients, with a history of substance use for which they are seeking treatment. Determination into group 1 (inhalants) or group 2 (other substance use) will be based upon the primary substance of concern, with no exclusion based on poly-drug use. Age, sex and community matched adolescents who have no history of substance use will be recruited to act as control participants, through either participant nomination or in collaboration with local youth groups. Participation will be voluntary and require informed consent, including parent/guardian consent for participants aged under 18 years.

9.2 Exclusion criteria Exclusion criteria include being unable to provide informed consent, participation considered harmful by the ADAWS team, or non-adherence. Furthermore, individuals who have been previously (i.e. prior to drug misuse) diagnosed with type 1 or type 2 diabetes will be excluded from the study. This is due to the fact that the study will measure levels of appetite regulating hormones such as insulin, and a history of diabetes and / or medications for managing diabetes will confound the study results. Pregnant women will be excluded from the study as pregnancy can alter metrics of body composition, growth, food intake, and hormone levels, which would confound the results of this study.

9.3 Potential for risk, burdens and benefits to participants The primary risk associated with participation in this study will be associated with blood collection, which can cause minor bruising or discomfort. This part of the study will be covered under a separate consent, so that participants can choose to answer the questionnaire and undergo body composition measurements, but decline to have blood Page 10 of 20 Inhalants and growth Dr Jhodie Duncan < AU/1/1F86217> < MR-2016-89> Protocol Version 1.1 Date: 21/07/2016 collected. To minimize this possibility blood will be collected by trained Mater health services staff. It is possible that the tests conducted could reveal that participants are at an increased risk for a previously undiagnosed health concern. If this should occur, participants will be appropriately referred to other health professionals within the Mater Health Service. ADAWS has policies and procedures in place for protecting the privacy and data of its clients, which will encompass data collected as part of this study. De-identified data will also be held by the Florey Institute, which also has approved policies for maintaining data confidentiality.

The benefits of this study will accrue to society in general, rather than to the participants of this study.

10. STUDY OUTCOMES

10.1 Primary Outcomes The primary outcomes are: 1. Height (measured in cm) 2. Body weight (measured in kg)

10.2 Secondary Outcomes Secondary outcomes are: 1. Demographic data (measured by a questionnaire) 2. Drug use history (measured by a questionnaire) 3. Food intake frequency (as a proxy for total dietary intake) (measured by a questionnaire) 4. Food preferences (measured by a questionnaire) 5. Adiposity (measured by waist circumference and skinfold tests) 6. Feelings of hunger (measured by questionnaire) 7. Levels of circulating appetite regulating hormones (measured from blood samples) 8. Glucose regulation (measured by a glucose tolerance test)

11. STUDY PROCEDURES

11.1 Recruitment and consent of participants Potential participants for the inhalants and other drugs study groups (groups 1 and 2) who meet the inclusion criteria will be identified by ADAWS staff at the time of admission and invited to participate in the study. Potential participants will be provided with the Information Sheet and Consent Form and a staff member will verbally explain the risks and benefits associated with the study. At this time, the ADAWS staff member will verbally reiterate that the standard of care an individual receives as part of their substance use treatment will not be affected by their decision on participation in this study.

If individuals choose to participate, they will be asked to sign a Consent Form, with a separate consent section for the blood collection. As the inclusion age is from 12-20, potential participants aged under 18 years will require consent

Page 11 of 20 Inhalants and growth Dr Jhodie Duncan < AU/1/1F86217> < MR-2016-89> Protocol Version 1.1 Date: 21/07/2016 from both a parent / guardian and the participant. There is a separate Consent Form for participants requiring parent / guardian consent. Consent forms have been developed in accordance with NHMRC guidelines.

Based on ADAWS historical data, approximately 24 people per year are admitted due to a primary inhalant use issue. Assuming a conservative 50% recruitment rate, over 18 months, we estimate that the primary study group will have approximately 20 participants. Thus, the total number of participants will be approximately 60 across the three groups.

Control group participants will be sought by approaching youth groups and church groups within comparable socio- economic regions to the study group. Participants may also nominate individuals, e.g. friends or siblings, to act as controls for the study. An Expression of Interest document will be sent to groups, with a request that interested individuals contact the study coordinator. Interested individuals will then be provided with an Information Sheet and Consent Form and will have the risks and benefits verbally explained to them. Control participants will also require parent / guardian consent if they are under 18 years of age.

11.2 Withdrawal of participants from a study A Withdrawal of Consent form has been developed and will be used if participants or their parents / guardians wish them to withdraw from the study.

11.2.1 Participant withdrawal from study procedures If an individual consents to participate in the study, but withdraws their consent for blood collection/GTT, then the participant data collected up to the time of withdrawal from the study procedures will still be considered in the data analysis. This will be explained in the participant information sheet.

11.2.2 Participant withdrawal from a study Data collected on study participants up to the time of withdrawal will remain in the study database in order for the study to be scientifically valid. This will be explained in the participant information sheet.

11.3 Randomisation Because participants will be assigned to groups based on their substance use history, it is not possible to randomise this aspect of the study. Study group participants will be clients of ADAWS and therefore neither the investigator nor participants will be blinded to group assignment. Participants will be allocated an identification code, therefore, the investigators conducting the data analysis and reporting will be blinded to the participants’ identities.

11.4 Measurement tools used Four instruments will be used for data collection: 1. A questionnaire that has been designed for this study, to collect information on drug use history and food intake 2. Direct measurement of body growth and composition (height, weight, waist circumference, skin fold tests for adiposity), which will be entered by an ADAWS staff member on to the questionnaire

Page 12 of 20 Inhalants and growth Dr Jhodie Duncan < AU/1/1F86217> < MR-2016-89> Protocol Version 1.1 Date: 21/07/2016

3. Review of childhood patient health records (from maternal child health ‘red book’) (if available) to determine a participant’s historic growth trajectory 4. Blood specimen collection, which includes the GTT

It is important for this study that a detailed drug use history is taken. An unpublished study (manuscript in review) by the lead and associate investigator found that height impairments associated with adolescent inhalant abuse are affected by the duration of inhalant use. Additionally, as the study is investigating substance abuse that is occurring during the adolescent growth spurt, it will be important to determine when substance use began. Lastly, different substances of abuse can have markedly different effects on body weight and food intake [37], thus it is important to collect data on each type of substance separately.

Food and drink intake data will be collected by a questionnaire, where participants are asked to reflect on food and drink intake from the prior two days. Whilst best practice would be a detailed food diary, this was deemed to be unfeasible for this study and likely to yield unreliable results, given the age and substance use concerns of these participants. Instead, a food and drink frequency questionnaire has been developed by the study team, to investigate how often different types of food are eaten, dietary composition, whether caloric intake is dominated by food or drink, the typical meal size, and food preferences. These are all important components as inhalant use has been shown to not only affect food intake, but also food preferences. A question regarding feelings of hunger when under the influence of substances of concern is included in order to determine if hunger is acutely affected by substance use. Lastly, a question relating to feelings of hunger is included, in order to provide a potential behavioural link to the levels of appetite-regulating hormones from the blood tests. Examples for meal sizes and meal types have been provided in the questionnaire, to reduce inter-participant variability in interpretation of the categories and thus improve data quality.

Growth and body composition will be assessed by direct measurement of height, weight, waist circumference, and skin fold tests for adiposity. These are all simple and standard growth measures, which will allow comparison to population growth percentiles. Body mass index (BMI) will be calculated from height and weight data. Whether an individual meets FTT criteria will be determined by comparison of height and weight data to population growth percentiles. Standard FTT criteria will be used; weight below the 3rd percentile or 20% below ideal weight for height.

Blood tests including the GTT will be undertaken by a trained nurse, in conjunction with other standard blood tests used as part of the ADAWS in-patient health check. Appetite-regulating hormone levels are standard tests and are able to analysed by Mater Pathology, in accordance with standard protocols. A glucose tolerance test requires consumption of a sugary liquid at a set period prior to the blood test.

All data will be collected by trained ADAWS staff members, who are experienced in collecting drug use history and general health data. A specific instruction sheet on taking skin fold measurements for adiposity will be provided. Given

Page 13 of 20 Inhalants and growth Dr Jhodie Duncan < AU/1/1F86217> < MR-2016-89> Protocol Version 1.1 Date: 21/07/2016 the simplicity of the growth measures and the standardised questionnaire, there is no intention to assess inter-rater variability.

11.5 Study involvement by participants Once participants have signed the consent form, all data collection will take place in appointments, which are integrated into the ADAWS health check. It is anticipated that data collection for physical measurements and questionnaire for this study will take less than 1 hour, for each participant. The blood test will be undertaken at the time of the health check, though not within the same appointment, as per current ADAWS procedures. The glucose tolerance test will require the consumption of a sugary liquid at a set time prior to the blood test.

This study is non-interventional and there is no follow-up, therefore, participants’ will not be monitored in relation to this study after completing the questionnaire and blood test. However, group 1 and 2 participants will continue to be monitored as clients of ADAWS.

All participants will be offered a $30 movie voucher as a gratuity for their participation.

11.6 Data management and storage Data will be collected in a confidential setting by ADAWS staff on the standard questionnaire, which will include a participant’s ID code with no identifying data. Participants will be asked to complete the demographics, drug use history, and food intake questions themselves in the presence of ADAWS staff. Upon request or if there are literacy issues, ADAWS staff may enter verbal responses from participants to the questionnaire. ADAWS staff will complete the body composition measures and record this data. Principal investigators at ADAWS will have access to a master participant sheet, which links an individual’s name to their study ID code, and this will not be provided to researchers at the Florey Institute or QUT. This document will be password protected and stored electronically on a network drive with restricted access. Both the paper questionnaire and the electronic data entry sheet (excel) will be de-identified and only the study ID code will be used for subsequent analysis that will be undertaken by a member of the study team blind to the original participant grouping. De-identified questionnaires and pathology forms will be provided to the Florey Institute, where results will be entered to an electronic data entry sheet. All members of the study team will have access to the de-identified data. Hardcopies of original questionnaires, consent forms, and pathology forms will be retained in locked drawers, at ADAWS. After transfer of de-identified information and copies of de-identified questionnaires to the Florey for data analysis, these copies will be retained in a locked drawer at the Florey.

Data confidentiality and security will be maintained by conforming to the Florey Privacy policy (201512) and Mater policy MHS-HRES-CRSU-1.05. Data will be retained securely for 33 years after the completion of the study.

When data is entered from the questionnaire to the spreadsheet, a number coding system will be used to allow for statistical analysis e.g. 1 for yes and 2 for no.

Page 14 of 20 Inhalants and growth Dr Jhodie Duncan < AU/1/1F86217> < MR-2016-89> Protocol Version 1.1 Date: 21/07/2016

11.7 Safety considerations/Patient safety This study has been designed to prioritise participant safety. Blood collection has the potential for minor discomfort and bruising, for which participants will receive standard information at the time of blood collection (e.g. short-term restrictions on lifting to minimise bruising). This risk is also covered in the Information Sheet, so that participants are appropriately informed. The questionnaire will be administered by trained ADAWS staff, so that any feelings of distress/anxiety from participants can be addressed immediately.

Reporting of adverse incidents or serious adverse incidents will be undertaken in accordance with the existing Mater policy.

11.8 Data monitoring

This study will be monitored by the Mater Research Oversight Committee, in accordance with existing policies and processes.

The following information will be provided to the Mater HREC:

 An annual progress report due on the anniversary of the approval date will be submitted to the Mater HREC and relevant Governance Office  A request for amendment will be applied for if:  new investigators are appointed  changes are being made to the study including the approved documentation

12. SAMPLE SIZE AND DATA ANALYSIS

12.1 Sample size and statistical power The power calculation for the primary outcomes is shown in Figure 1. This power calculation is based on unpublished data (manuscript in review) from the study group, which investigated height and weight changes associated with adolescent petrol sniffing (a form of inhalant abuse). The effect size was calculated as 0.38, based upon this data. Assuming a Repeated Measure ANOVA for height and weight (two variables), with a standard type 1 error of 0.05 and a conservative type 2 error of 0.90, across three groups, the required total sample size is 69. Thus, this matches with our estimated sample size of approximately 60 participants.

Page 15 of 20 Inhalants and growth Dr Jhodie Duncan < AU/1/1F86217> < MR-2016-89> Protocol Version 1.1 Date: 21/07/2016

Figure 1 - power calculation for primary outcomes

12.2 Data analysis plan As this study is not randomised, an Intention to Treat analysis will not be conducted. Participants who only provide partial data via the questionnaire will still be included in the study. Any deviation to the data collection or statistical plan will be addressed via an ethics amendment request. Statistical analysis will include the following tests, with guidance on analysis and post-hoc tests provided by the Florey’s Statistical Consulting Core Service:  Repeated measures ANOVA will be used for dependent variables (e.g. height and weight).  One-way ANOVA (by group) will be used for independent variables (e.g. food intake as measured by a total food frequency score or body mass index).  Chi-square tests will be used for categorical variables (e.g. does the participant smoke cigarettes; yes or no, or does the participant meet FTT criteria; yes or no).

13. ETHICAL CONSIDERATIONS Ethics approval for this study will be sought from the Mater Human Research Ethics Committee, which is NHMRC certified and able to approve multi-centre research studies with paediatric and adult participants. The study will be conducted in full conformance with the principles of the National Statement on Ethical Conduct in Human Research, and all other relevant guidance documents and within the laws and regulations of Australia.

14. DISSEMINATION OF RESULTS AND PUBLICATIONS Upon completion of the study, a summary of the project findings will be produced, and emailed to study participants. It is anticipated that the study findings will be published in a scientific journal. Authorship will be determined in

Page 16 of 20 Inhalants and growth Dr Jhodie Duncan < AU/1/1F86217> < MR-2016-89> Protocol Version 1.1 Date: 21/07/2016 accordance with the International Committee of Medical Journal Editors requirements for authorship; however, we anticipate that authorship will be: - Rose Crossin (Florey, PhD student and Coordinating Investigator) - Nicole Lacey (ADAWS, Local Coordinating Investigator) - Amanda Tilse (ADAWS, AI) - Greg McGahan (ADAWS) - Leanne Hides (QUT, AI) - Andrew Lawrence (Florey, PhD supervisor of Rose Crossin) - Jhodie Duncan (Florey, Principal Investigator) Rose Crossin will take the lead role in the publication of this data, in accordance with the requirements of her PhD. Upon request, study participants will be provided with a copy of the journal article upon publication. Other communication may be undertaken, including, but not limited to, media releases, conference presentations, and seminars.

15. OUTCOMES AND SIGNIFICANCE This study will be the first to attempt to characterise the effects of adolescent inhalant abuse on growth, food intake and metabolic control. The findings from this study will be used to guide treatment strategies for adolescents who have a history of inhalant use, in order to minimise the health risks (both during inhalant use and after inhalant use has ceased). For example, if inhalants were found to alter food intake or food preferences, treatment strategies may include nutritional counselling. Alternatively, if altered levels of hormones were found to contribute to alterations in food intake, this may lead to new therapies being developed including replacement therapies. The findings may also be used to inform young people about the potential health risks linked with using inhalants, by including any effects on growth in information sheets about inhalant abuse.

16. BUDGET The budget estimates assumes 60 participants; 40 of whom are ADAWS clients and 20 of whom are controls Purchase of skin fold callipers Used as a measure of adiposity $50 Plasma glucose and insulin These hormones contribute to appetite 60 x $140 = $8 400 regulation. These tests cannot be taken from a standard blood panel, therefore, they will need to be paid for separately Plasma leptin and ghrelin (for ADAWS These hormones contribute to appetite 40 x 0 = $0 participants) regulation. These can be added to the standard blood panel for ADAWS participants, so will not need to be paid for separately. If these cannot be added to the standard blood panel, any additional costs will be added to the

Page 17 of 20 Inhalants and growth Dr Jhodie Duncan < AU/1/1F86217> < MR-2016-89> Protocol Version 1.1 Date: 21/07/2016

research budget. Plasma leptin and ghrelin (for control These hormones contribute to appetite 20 x $100 = $2 000 participants) regulation. A gratuity for participation via movie The study will require a visit to the Mater 60 x $30 = $1 800 voucher for all participants Hospital, therefore, it is appropriate to provide a gratuity to participants Florey statistical core consulting service To provide advice and guidance on $200 statistical analysis Total costs $12 450

This project will be internally funded by the Florey Institute of Neuroscience and Mental Health, with in-kind support by ADAWS by the incorporation of this project into existing processes. Funding via project grants to support this project may be pursued as opportunities become available, at the discretion of the study team.

17. GLOSSARY OF ABBREVIATIONS ADAWS – Mater Adolescent Drug and Alcohol Withdrawal Service Florey – The Florey Institute of Neuroscience and Mental Health FTT – Failure to Thrive QUT – Queensland University of Technology GTT – Glucose Tolerance Test

18. REFERENCES 1. ADCA, Policy Position - Inhalants. 2010, Alcohol and other Drugs Council of Australia: Canberra. 2. VHS, National Directions on Inhalant Abuse. 2005, Victorian Department of Human Services: Melbourne. 3. Duncan, J.R. and A.J. Lawrence, Conventional concepts and new perspectives for understanding the addictive properties of inhalants. Journal of pharmacological sciences, 2013. 122(4): p. 237-243. 4. Funada, M., et al., Evaluation of rewarding effect of toluene by the conditioned place preference procedure in mice. Brain research protocols, 2002. 10(1): p. 47-54. 5. AIHW, Substance abuse among Aboriginal and Torres Strait Islander people. 2011, Australian Institute of Health and Welfare: Canberra. 6. Cairney, S., et al., The neurobehavioural consequences of petrol (gasoline) sniffing. Neuroscience & Biobehavioral Reviews, 2002. 26(1): p. 81-89. 7. AIHW, 2010 National Drug Strategy Household Survey Report. 2011, Australian Institute of Health and Welfare: Canberra. 8. CCV, Australian secondary school students' use of tobacco, alcohol, and over-the-counter and illicit substances in 2011. 2012, Cancer Council of Victoria for Australian Government Department for Health and Ageing: Melbourne.

Page 18 of 20 Inhalants and growth Dr Jhodie Duncan < AU/1/1F86217> < MR-2016-89> Protocol Version 1.1 Date: 21/07/2016

9. Dell, C.A., S.W. Gust, and S. MacLean, Global issues in volatile substance misuse. Substance use & misuse, 2011. 46(sup1): p. 1-7. 10. TAPIA‐CONYER, R., et al., Risk factors for inhalant abuse in juvenile offenders: the case of Mexico. Addiction, 1995. 90(1): p. 43-49. 11. Green, M., Bright Futures: Guidelines for Health Supervision of Infants, Children, and Adolescents. 1994: ERIC. 12. Landrigan, P.J. and A. Garg, Chronic effects of toxic environmental exposures on children's health. Journal of Toxicology: Clinical Toxicology, 2002. 40(4): p. 449-456. 13. Lubman, D.I., M. Yücel, and W.D. Hall, Substance use and the adolescent brain: a toxic combination? Journal of Psychopharmacology, 2007. 21(8): p. 792-794. 14. Tanner, J. and R. Whitehouse, Clinical longitudinal standards for height, weight, height velocity, weight velocity, and stages of puberty. Archives of disease in childhood, 1976. 51(3): p. 170-179. 15. BONJOUR, J.-P., et al., Critical Years and Stages of Puberty for Spinal and Femoral Bone Mass Accumulation during Adolescence*. The Journal of Clinical Endocrinology & Metabolism, 1991. 73(3): p. 555-563. 16. Gordon, C., Measurement of bone density in children. Curr Opin Endocrinol Metab, 2005. 12: p. 444-51. 17. Dreizen, S., C.N. Spirakis, and R.E. Stone, A comparison of skeletal growth and maturation in undernourished and well-nourished girls before and after menarche. The Journal of pediatrics, 1967. 70(2): p. 256-263. 18. Martorell, R., L.K. Khan, and D.G. Schroeder, Reversibility of stunting: epidemiological findings in children from developing countries. European journal of clinical nutrition, 1994. 48: p. S45-57. 19. Reed, R.B. and H.C. Stuart, Patterns of growth in height and weight from birth to eighteen years of age. Pediatrics, 1959. 24(5): p. 904-921. 20. Pisetsky, E.M., et al., Disordered eating and substance use in high‐school students: Results from the Youth Risk Behavior Surveillance System. International Journal of Eating Disorders, 2008. 41(5): p. 464-470. 21. Glaser, H.H. and O.N. Massengale, Glue-sniffing in children: Deliberate inhalation of vaporized plastic cements. JAMA, 1962. 181(4): p. 300-303. 22. Ryu, Y.H., et al., Cerebral perfusion impairment in a patient with toluene abuse. Journal of nuclear medicine: official publication, Society of Nuclear Medicine, 1998. 39(4): p. 632-633. 23. Morón, L., et al., Toluene alters appetite, NPY, and galanin immunostaining in the rat hypothalamus. Neurotoxicology and teratology, 2004. 26(2): p. 195-200. 24. Jarosz, P.A., et al., Effects of abuse pattern of gestational toluene exposure on metabolism, feeding and body composition. Physiology & behavior, 2008. 93(4): p. 984-993. 25. Duncan, J.R., et al., Adolescent toluene inhalation in rats affects white matter maturation with the potential for recovery following abstinence. PloS one, 2012. 7(9): p. e44790. 26. Wang, D.-H., et al., Reduced serum levels of ALT and GGT and high carbohydrate intake among workers exposed to toluene below the threshold limit values. Industrial health, 1998. 36(1): p. 14-19. 27. Dick, A., Chronic intermittent toluene inhalation in adolescent rats results in long lasting metabolic dysfunction with altered glucose homeostasis, in Psychoneuroendocrinology. 2015.

Page 19 of 20 Inhalants and growth Dr Jhodie Duncan < AU/1/1F86217> < MR-2016-89> Protocol Version 1.1 Date: 21/07/2016

28. Jackson, C.E., et al., Hunger and the perception of the scent of petrol: A potential neurobiological basis for increased risk of petrol inhalation abuse. Addiction Research & Theory, 2009. 17(5): p. 518-524. 29. Brady, M., Heavy metal: the social meaning of petrol sniffing in Australia. 1992: Aboriginal studies press. 30. Pryor, G.T., A toluene-induced motor syndrome in rats resembling that seen in some human solvent abusers. Neurotoxicology and teratology, 1991. 13(4): p. 387-400. 31. Roberts, L., A. Bevans, and C. Schreiner, Developmental and reproductive toxicity evaluation of toluene vapor in the rat: I. Reproductive toxicity. Reproductive Toxicology, 2003. 17(6): p. 649-658. 32. Pearson, M.A., et al., Toluene embryopathy: delineation of the phenotype and comparison with fetal alcohol syndrome. Pediatrics, 1994. 93(2): p. 211-215. 33. Malik, V.S., et al., Sugar-sweetened beverages and risk of metabolic syndrome and type 2 diabetes A meta- analysis. Diabetes care, 2010. 33(11): p. 2477-2483. 34. Hu, F.B., et al., Diet, lifestyle, and the risk of type 2 diabetes mellitus in women. New England Journal of Medicine, 2001. 345(11): p. 790-797. 35. Won, Y.L., et al., The Effects of Long-Term, Low-Level Exposure to Monocyclic Aromatic Hydrocarbons on Worker's Insulin Resistance. Safety and health at work, 2011. 2(4): p. 365-374. 36. Grundy, S.M., et al., Definition of metabolic syndrome report of the National Heart, Lung, and Blood Institute/American Heart Association Conference on scientific issues related to definition. Circulation, 2004. 109(3): p. 433-438. 37. Nolan, L.J., Shared urges? The links between drugs of abuse, eating, and body weight. Current Obesity Reports, 2013. 2(2): p. 150-156.

Page 20 of 20 Inhalants and growth Dr Jhodie Duncan < AU/1/1F86217> < MR-2016-89> Protocol Version 1.1 Date: 21/07/2016

Rose Crossin (737900)

Appendix 4 – Questionnaire for the human cohort study The approved research questionnaire is appended for additional information to the human cohort study described in Appendix 2.

183

Project title: The effect of adolescent inhalant use on growth and food intake

Patient ID code (as allocated by Participant Information Master sheet) : Date of exam: Person conducting exam:

Section 1 – background information

Date of birth:

Race/ethnicity:

Sex: Male Female Intersex

Highest level of education: Still in school Year 10 Year 12 TAFE University

Employment status: Still in school Studying Unemployed Part-time Full-time

List of current medications:

Have you ever been diagnosed with type 1 or type 2 diabetes: Yes No

Section 2 – body composition (to be measured during appointment)

Current height (cm): Skinfolds

Current weight (kg): Bicep:

Waist circumference (cm): Tricep:

Age at most recent child health check: Subscapula: (From child health record) Height from most recent child health check: Suprailiac: (From child health record) Weight from most recent child health check: (From child health record)

Inhalants and growth questionnaire master 21/07/2016 version 1.1 Page 1 of 6

Section 3 – drug use

3.1 What is your primary substance of concern / what substance is worrying you most:

3.1(a). If inhalants are the primary substance of concern, what products do you most commonly sniff? (e.g. glue, petrol, thinners, etc):

Drug use history:

Please only complete one column for each drug (never used, used in the past, or still use). Greyed out boxes don’t require an answer.

For drugs where there is a severity score: 1=light use (e.g. one or two alcoholic drinks), 2=moderate use (e.g. three or four alcoholic drinks), 3=heavy use (e.g. five or more alcoholic drinks at one time)

Never used Used in the past Still use 3.2 Alcohol At what age did you start? At what age did you stop using this? Frequency Occasional of use Monthly Weekly Daily Severity score (1, 2, 3) 3.3 Cigarettes At what age did you start? At what age did you stop using this? Frequency Occasional of use Monthly Weekly Daily Severity score (1, 2, 3) 3.4 Inhalants At what age did you start? At what age did you stop using this? Frequency Occasional of use Monthly Weekly Daily Severity score (1, 2, 3)

Inhalants and growth questionnaire master 21/07/2016 version 1.1 Page 2 of 6

Never used Used in the past Still use 3.5 Marijuana At what age did you start? At what age did you stop using this? Frequency Occasional of use Monthly Weekly Daily Severity score (1, 2, 3) 3.6 Heroin At what age did you start? At what age did you stop using this? Frequency Occasional of use Monthly Weekly Daily 3.7 Cocaine At what age did you start? At what age did you stop using this? Frequency Occasional of use Monthly Weekly Daily 3.8 Methamphetamines / ice At what age did you start? At what age did you stop using this? Frequency Occasional of use Monthly Weekly Daily 3.9 Prescription drugs (illicit use) At what age did you start? At what age did you stop using this? Frequency Occasional of use Monthly Weekly Daily 3.10 Ecstasy / MDMA At what age did you start? At what age did you stop using this? Frequency Occasional Inhalants and growth questionnaire master 21/07/2016 version 1.1 Page 3 of 6

Never used Used in the past Still use of use Monthly Weekly Daily 3.11 Other: At what age did you start? At what age did you stop using this? Frequency Occasional of use Monthly Weekly Daily

Inhalants and growth questionnaire master 21/07/2016 version 1.1 Page 4 of 6

Section 4 – diet and food preferences

4.1 How much do you typically eat in a day? (please tick one box for each meal)

Don’t eat Small amount Moderate amount Large amount On a typical day, I Would fit on a Would fit on a Would not fit on a wouldn’t eat small plate / bowl dinner plate dinner plate anything for this E.g. a piece of E.g. pasta and E.g. a burger with meal toast, 1-2 biscuits sauce, a filled roll fries followed by dessert Breakfast

Lunch

Dinner

Snacks

4.2 In the last 2 days, please estimate how many times have you eaten:

Whole fruit Bread Ice cream

Vegetables Pasta or rice Biscuits

Meat or fish Breakfast cereal Potato crisps

Dairy products Sweets Fried food (e.g. cheese / yoghurt) (e.g. lollies / chocolate) (e.g. chicken wings) Eggs Cakes / pastries Fast food / take away (e.g. pizza / burgers)

4.3 In the last 2 days, please estimate how many times have you drunk:

Water Fruit juice Milk

Soft drinks Energy drinks Tea / coffee

Inhalants and growth questionnaire master 21/07/2016 version 1.1 Page 5 of 6

4.4 Which of these foods do you find the most appealing? (tick only 1)

A B C D

4.5 After you have used your primary drug of concern (within 12 hours of drug use), what are your feelings towards food / eating? (tick only 1)

Don’t feel hungry

Feel hungry, but don’t actually end up eating Craving particular foods

High levels of hunger and food consumption

4.6 If you don’t eat anything for 3-4 hours, how do you feel? (tick as many as apply)

Feel fine / don’t feel hungry

Feeling of emptiness in stomach

Craving particular foods

Faint / lightheaded

Grumpy / irritable

Lethargic / low energy

This is the end of the questionnaire – thank you for your time.

Inhalants and growth questionnaire master 21/07/2016 version 1.1 Page 6 of 6

Minerva Access is the Institutional Repository of The University of Melbourne

Author/s: Crossin, Rose

Title: The growth and energy balance consequences of adolescent inhalant abuse

Date: 2018

Persistent Link: http://hdl.handle.net/11343/216872

File Description: The growth and energy balance consequences of adolescent inhalant abuse

Terms and Conditions: Terms and Conditions: Copyright in works deposited in Minerva Access is retained by the copyright owner. The work may not be altered without permission from the copyright owner. Readers may only download, print and save electronic copies of whole works for their own personal non-commercial use. Any use that exceeds these limits requires permission from the copyright owner. Attribution is essential when quoting or paraphrasing from these works.