Skip to main content
Integrative and Comparative Biology logoLink to Integrative and Comparative Biology
. 2018 Oct 26;58(6):1043–1054. doi: 10.1093/icb/icy118

Two Decades of the Impact of Tasmanian Devil Facial Tumor Disease

Gregory M Woods 1,, Samantha Fox 2,3, Andrew S Flies 1, Cesar D Tovar 1,4, Menna Jones 5, Rodrigo Hamede 5, David Pemberton 2, A Bruce Lyons 4, Silvana S Bettiol 4
PMCID: PMC6927850  PMID: 30252058

Abstract

The Tasmanian devil, a marsupial carnivore, has been restricted to the island state of Tasmania since its extinction on the Australian mainland about 3000 years ago. In the past two decades, this species has experienced severe population decline due to the emergence of devil facial tumor disease (DFTD), a transmissible cancer. During these 20 years, scientists have puzzled over the immunological and evolutionary responses by the Tasmanian devil to this transmissible cancer. Targeted strategies in population management and disease control have been developed as well as comparative processes to identify variation in tumor and host genetics. A multi-disciplinary approach with multi-institutional teams has produced considerable advances over the last decade. This has led to a greater understanding of the molecular pathogenesis and genomic classification of this cancer. New and promising developments in the Tasmanian devil’s story include evidence that most immunized, and some wild devils, can produce an immune response to DFTD. Furthermore, epidemiology combined with genomic studies suggest a rapid evolution to the disease and that DFTD will become an endemic disease. Since 1998 there have been more than 350 publications, distributed over 37 Web of Science categories. A unique endemic island species has become an international curiosity that is in the spotlight of integrative and comparative biology research.

Introduction to the Tasmanian devil and devil facial tumor disease

The Tasmanian devil (Sarcophilus harrisii), is a member of the Dasyuridae family and is the world’s largest carnivorous marsupial, now endemic to the island of Tasmania, Australia. The devil is most commonly found along Tasmania’s eastern half, and northern and northwest coastline in dry forests, woodlands, and coastal scrub (Jones and Barmuta 2000). Devils are the top predator in Tasmanian ecosystems (Hollings et al. 2016) and the dominant scavenger with morphological specializations for consuming bone (Jones 2003). Devils are primarily nocturnal, with a home range in the order of 4–27 km2 and a mean daily movement of 9 km (Pemberton 1990). The social behavior of sexual contact and competition for food leads to fighting which is characterized by biting, with many of the bites occurring on the face (Hamede et al. 2008).

Tasmanian devils have been isolated in Tasmania for approximately 14,000 years after sea level rise created Bass Strait (Lambeck and Chappell 2001). Devils are now considered endemic to Tasmania following their extinction on the Australian mainland about 3000 years ago (Bruniche-Olsen et al. 2018; White et al. 2018). The extinction was likely driven by El Niño Southern Oscillation (ENSO)-related climate change with additional direct and indirect pressures from human intensification (Johnson and Wroe 2003; Prowse et al. 2014) and the introduction of dingoes (Letnic et al. 2012). Guiler (1982) reported very low populations of devils in the first half of the 1900s, coinciding with the population crash of the thylacine.

The emergence of the fatal transmissible cancer, DFTD, in 1996 at Waterhouse Point in north-east Tasmania has caused an overall decline in the devil population by approximately 80% (Lazenby et al. 2018). Local declines of up to 95% have been recorded (McCallum et al. 2007; Lazenby et al. 2018). The first case of a second, and independent, transmissible cancer was detected in south-east Tasmania in 2014 (Pye et al. 2016b). Consequently, DFTD comprises two independent transmissible cancers, DFT1 (first identified in 1996) and DFT2 (first identified in 2014). DFT2 is currently restricted to the southeast of the island whereas DFT1 is spread across most of mainland Tasmania (Fig. 1). Both DFTs cause tumors primarily on the face, neck, and oral cavity of the Tasmanian devil, and are first visible as small nodules on mucosal and dermal surfaces. DFT1 originated in a Schwann cell of a female devil (Murchison et al. 2010) whereas DFT2 arose in a male devil, presumably of a similar cellular origin to DFT1 (Pye et al. 2016b; Stammnitz et al. 2018). Nearly all animals diagnosed with the tumor have been recorded to succumb to the disease. Throughout this review DFTD refers to the disease caused by either DFT1 or DFT2. The emphasis will be on DFT1. Tasmanian devils have been formally listed as an endangered species since 2008 by the International Union for Conservation of Nature (IUCN) and they are also protected by both State and Federal legislation (IUCN Red List).

Fig. 1.

Fig. 1

Spread of DFTD. DFT1 was first recorded in the north-east Tasmania (green) in 1996. The first recorded case of DFT2 was separated by distance and time to DFT1 as the first case of DFT2 (red) was in 2014 in south east Tasmania. DFT1 has spread over most of the island, whereas DFT2 is contained to a small area.

Comparative biological approaches to identify DFTD

A primary limitation to understanding the interaction between the devil immune system and DFTD cells has been the lack of species-specific tools, a problem that continues to pervade comparative immunology research on non-traditional study species (Brock et al. 2014). White blood cell (WBC) counts can be done without species-specific reagents and thus are often used as a diagnostic test or general measure of immune function in many species (Nunn et al. 2003; Bordes and Morand 2009; Cooper et al. 2012). Analyses of hematological and serum biochemical data are also generalized methods to probe basic immunological parameters. Such methods have been used for marsupials, including Tasmanian devils (Parsons et al. 1971; Peck et al. 2015).

Immunoglobulins play an important role in many aspects of immune function, but their role in cancer remains ambiguous even in species where isotypes, Fc receptors, and functions have been thoroughly characterized (Nimmerjahn and Ravetch 2007). Defining the role of immunoglobulins in DFTD, and in wild animals in general, is a critical task. Few studies have found strong correlations between survival and pathogen-specific antibodies (Nussey et al. 2014), but in general immunoglobulin levels in serum remain a blunt tool for predicting infection status.

Basic lymphoid tissue anatomy has provided solid, although relatively basic, morphological information. Immunohistochemistry has provided a valuable tool in advancing knowledge of lymphoid architecture. Cross-reacting antibodies developed to investigate other species (e.g., human and mouse) have yielded information about immune–pathogen interplay in non-traditional species (Jurd 1994; Coutinho et al. 1995; Hemsley et al. 1995; Canfield and Hemsley 2000).

Histology and immunohistochemistry using cross-reactive antibodies have been useful tools to investigate the DFTD tumor and the major immunological structural components of the Tasmanian devil (Loh et al. 2006a, 2006b). Initial studies mapped DFTD (DFT1) as an undifferentiated soft tissue neoplasm (Loh et al. 2006a). Immunohistochemistry, with cross-reacting antibodies suggested that DFTD (DFT1) was of neuroectodermal origin (Loh et al. 2006b). Transcriptome sequencing, which is relevant for any species, revealed the original DFTD (DFT1) to have been derived from a Schwann cell (Murchison et al. 2012). Chromosome painting and karyotyping, techniques adaptable to all species, determined that chromosomes 1 and X contain most of the chromosomal rearrangements (Deakin et al. 2012). A major diagnostic breakthrough that provided a valuable research tool became available when a cross reactive anti-periaxin antibody was found to be a reliable marker to identify DFT1 cells (Tovar et al. 2011).

Monitoring the spread of DFTD

Although it was recognized that Dasyurids, and specifically Tasmanian devils, commonly develop a variety of neoplasms (Griner 1979; Canfield and Cunningham 1993) data indicated that the earliest report of a facial tumor was in 1996 (Hawkins et al. 2006). Targeted monitoring for the original DFTD (DFT1) was first performed in 2003 to determine how much of Tasmania was affected by this devastating but little known disease. An informal examination of trapped devils in a state-wide “snapshot” survey showed DFTD (DFT1) to be present in approximately one-third of devil populations surveyed (N. Mooney, personal communication). By 2005, histological confirmation of DFTD (DFT1) in 41 sites covered 51% of the area of devil occurrence in Tasmania (Hawkins et al. 2006). Monitoring of devil populations over the following 13 years showed DFTD (DFT1) to have spread in a south-westerly direction to cover approximately 80% of the state. The rate of spread was affected by geography as well as movement patterns and abundance of devils (Lazenby et al. 2018). In one relatively abundant population in the north-west of the state, where DFTD (DFT1) had arrived relatively recently, repeated monitoring over 3 years showed DFTD (DFT1) to be spreading at approximately 5–7 km per year (Save the Tasmanian Devil Program [STDP], Tasmanian State Government, unpublished data).

Two principle methods have been employed to monitor the spread of DFTD (DFT1). Devils were trapped in specially designed tubular traps made from PVC pipe (diameter 315 mm × length 875 mm; N. Mooney and D. Ralph, unpublished data). The trapping was performed at the most western edge of the known location of DFTD (DFT1), and was carried out annually (2009–2013). Several teams were used to cover a large area in the north-west of the state. As cases of DFTD (DFT1) were located at the disease “front,” the trapping teams steadily moved their trap lines west until they no longer recorded any cases of disease. This method relied on histological confirmation of DFTD (DFT1), recorded the change in age distribution and abundance of the populations following disease arrival, and mapped the rate of spread of the disease. For many of these infected areas, the annual monitoring precluded specific time of disease arrival. This was particularly relevant when trapping teams moved into areas of very recent DFTD (DFT1) arrival. With very low disease prevalence, finding cases of DFT1 was either a matter of enormous resourcing or serendipity.

The second method of monitoring takes advantage of the more recent advances in motion sensing cameras (Thalmann et al. 2014; Meek et al. 2015). Cameras are set to cover a certain area to give year-round monitoring of disease presence. Cameras are set with meat baits and lure canisters, which along with memory cards and batteries, need to be changed every 2 months. Visitation by devils to the cameras reduces as the meat is consumed and the lure canisters dry out and lose their smell. The biggest challenge in analyzing images from the cameras is distinguishing between scar tissue from old wounds, fresh lumpy wounds, and a DFTD tumor. Sometimes this can be impossible to discern, especially if the tumor is inside the oral cavity. Without having the animal in hand there is no ability to feel the lump, which if DFT1 will have a very hard base to it. The advantage of cameras over trapping is the likelihood of seeing more devils. Reports to the STDP through roadkill sightings or the hotline used by the general public have proved effective in tracking occurrences of DFTD. New cases of DFT1 were located further west of the disease-front than previously recorded and the first case of DFT2 was a result of the hotline.

Movement of diseased devils is the principal determinant of the rate of spread of DFTD, and a number of factors including geography, abundance of devils, and food availability are likely to play important roles in determining movement. Rough estimates of disease spread can be based on the decline in spotlight counts of devils combined with confirmed cases of the disease. The decline in spotlight counts provides an indication of loss of devils and the need to confirm cases validates that the population decline was due to DFTD (DFT1). The spread from the north to the south of the state has been more rapid than that from the east to the west (McCallum et al. 2007). Accurate estimates of the rate of disease spread are challenging because only devils with visible symptoms of DFTD can be diagnosed. Not all devils within a given area are sampled during any one survey (Bode et al. 2009), although the recapture probability of devils is high (in the order of 80%) (Lachish et al. 2007). Imperfect detection is a challenge common to surveys of wild animals (Williams et al. 2002). The rate of spread of the disease is variable as different barriers (rivers, mountains, dense bush) affect devil movements (Storfer et al. 2017). The rate of spread appears to vary between 7 and 52 km/year (McCallum et al. 2007).

Evidence that DFTD (DFT1) is transmissible (contagious)

A remarkable feature of DFTD (DFT1) is that the cancer cell itself is transmitted between unrelated devils as an infectious “pathogen.” As a contagious cancer, the malignant cells are transferred between individuals as a natural allograft, forming new clonal tumors reviewed in Woods et al. (2015). The original proposal for the “allograft theory of transmission” came from basic studies of the karyotype of the DFTD (DFT1) cells (Pearse and Swift 2006). Chromosomes of the cancer cells had complex chromosomal rearrangements. However, the cancer cells from devils from different locations showed an identical karyotype, which in every case was different from the host. More detailed karyotypic studies confirmed that DFTD (DFT1) cells from different devils have similar complex chromosomal rearrangement (Deakin and Belov 2012; Murchison et al. 2012; Pearse et al. 2012). Genetic studies of multiple DFT1 tumors at the DNA, microsatellite, and major histocompatibility complex loci all provide support for the clonal nature of the cancer cells and therefore the contagious nature of the tumor (Siddle et al. 2007; Murchison et al. 2010, 2012). The genomes of DFTD (DFT1) tumors on two devils isolated in distance and time shared an almost identical genetic variation (Murchison et al. 2012). Genome sequencing of DFTD (DFT1) tumors identified that all DFT1s share point variants, structural variants, and copy number changes, which are distinct from their hosts (Miller et al. 2011; Murchison et al. 2012). Although there was evidence of karyotypic evolution, the DFTD (DFT1) genome appeared stable, hence the long term survival of this transmissible cancer. DFTD (DFT1) distinguishes itself from other cancers, not only just due to transmissibility, but also genomic stability which is not found in most solid tumors (Hanahan and Weinberg 2000, 2011). The canine transmissible venereal tumor has also demonstrated relative genomic stability across 11,000 years of inter-individual transmission (Strakova and Murchison 2015), suggesting the selection pressure maintains key functional genes across time and individuals.

Immune system of the Tasmanian devil

As allografts produce strong immune responses, “grafted” DFTD (DFT1) cancer cells had to overcome immunological barriers of the host (Woods et al. 2007). Either the immune system of the host or the nature of the cancer cells could have contributed to the failure to reject the “grafted” DFT1 cells. At the time of DFTD (DFT1) discovery, very little was known about the immune system of the Tasmanian devil. Consequently, and given the lack of reagents to probe the devil’s immune system, initial research relied on basic analyses of blood films and lymphoid organs. This was especially relevant as marsupials were believed to have a “weaker” immune response than that of placental mammals (Jurd 1994).

Differential WBC counts and lymphoid organ analyses were similar in appearance to those in placental mammals (Kreiss et al. 2008, 2009a). An analysis of serum biochemistry and hematology against DFTD (DFT1) status revealed that total WBC counts and neutrophils were higher in diseased devils, whereas lymphocytes were decreased; this suggests that the devil immune system is affected by DFTD in a manner similar to an acute infection and that WBC levels are predictive of disease status (Peck et al. 2016). Interestingly, this study found no association between disease status and serum immunoglobulin levels, another routine diagnostic test that can be predictive of acute infection status.

The development of specific monoclonal antibodies was required for the identification of lymphocyte subsets. CD4+ and CD8+ T cells were identified in lymphoid organs and as seen in lymphoid tissues of other mammals, there were more CD4+ than CD8+ cells (Howson et al. 2014). Functional assessment of lymphocyte proliferation, phagocytosis, skin graft rejections, antibody responses to immunizations with sheep red blood cells, and cytotoxicity assays for innate killer cells such as NK-like cells demonstrate that there was no evidence for immune defects (Kreiss et al. 2008, 2009b, 2011a, 2015; Brown et al. 2011, 2016). As Tasmanian devils scavenge on carcasses that would have high microbial loads, the ability to recognize pathogens would be paramount to survival. Consequently, innate cell receptors such as toll like receptors (TLRs) would need to be present and functional. This was indeed the case as the innate cells of the Tasmanian devil expressed a comprehensive range of functional TLRs (Patchett et al. 2015, 2017).

Immune response to DFTD (DFT1)

It is clear that healthy devils are not immunosuppressed and should respond to DFTD (DFT1) “allografts.” Tasmanian devils have a reduced genetic diversity, potentially due to a “founder effect” or a “bottleneck” in the population (Jones et al. 2004). This reduced diversity was particularly evident at the MHC gene loci, and this was the primary focus of most early studies to explain the lack of allogenic recognition of DFTD (Siddle et al. 2007). However, as skin grafts are rejected, limited MHC diversity may contribute to, but cannot fully explain, the lack of rejection of DFTD “grafts.” As cancer cells can downregulate MHC-I to make them evade the immune system (Fassati and Mitchison 2010) the expression of MHC-I was investigated. It was discovered that the DFTD (DFT1) cells do not express MHC-I antigens on their cell surfaces (Siddle et al. 2013). The absence of MHC expression is most likely the major cause for the failure to induce an allogeneic response thereby making the cancer cells effectively “invisible” to the devil’s immune system. Other contributing factors might include cytokines (Morris and Belov 2013), the loss of the original dendritic cells from the index case preventing direct recognition, down regulation of tumor-related molecules (e.g., DAMPS, TSTAs), up-regulation of inhibitory cell surface signaling molecules (e.g., PD-L1 (Flies et al. 2017); and the tumor microenvironment (Hui and Chen 2015).

A mouse xenograft model has also been developed for understanding tumor biology in a live host (Kreiss et al. 2011b; Pinfold et al. 2014). These studies provided the initial in vivo evidence that an immune response can be generated against the DFTD cells. The use of immunodeficient mice has provided researchers with a model that extends in vitro studies, without the need to use the endangered species under investigation. Although most of the studies of human diseases using immunodeficient mice has led to “humanized mice” (Ito et al. 2018), such a model could be adapted to non-classical species.

Epidemiology of DFTD and alteration of population structure

DFTD is almost 100% fatal, usually resulting in death within 9–12 months of the presentation of a tumor (Hamede et al. 2012) and potentially at least 2 years for some devils after initial inoculation (Wells et al. 2017). Unlike most infectious diseases, DFTD (DFT1) has its greatest effect on the “fittest” population, including devils with the highest reproductive output (Wells et al. 2017). Once DFTD (DFT1) has invaded a population, an important indicative sign of disease presence is the loss of the older age classes. In eastern Tasmania, where the disease originated, population growth rate declined by 50% per year during the first 6 years following disease outbreak (Lachish et al. 2007). The typical pattern is population decline with most of the remaining animals falling into the 1–2 year age class (Lazenby et al. 2018). Mature breeding individuals (2–4 years old; devils usually live for 5–6 years in the wild and rarely breed beyond the age of 5 years) disappear first from a population, followed by the 2 year olds (the usual age of sexual maturity in a DFTD-free population) (Lachish et al. 2009). This primary reduction in age class is probably due to disease transmission occurring during the mating season between sexually mature devils. As DFTD is spread by injurious contact when individuals bite each other, transmission is likely greatest among adults of breeding age as injuries from intraspecific bites peak during the mating season (Hamede et al. 2008). These seasonal and demographic patterns of biting injuries underlie the demographic changes of progressive reduction in age structure that follow disease outbreak (Lachish et al. 2009).

A consequence of the reduction in local population density following DFTD outbreak is that growth rates of sub-adult devils increase. A greater prey availability and nutrition enable a greater proportion of younger females to attain a sufficient body mass to breed in their sub-adult year (age 1 year) (Jones et al. 2008a; Lachish et al. 2009). This reduced competition has facilitated precocial breeding with females mating at the age of 1 year, rather than the usual 2 year sexual maturity point (Lachish et al. 2007; Jones et al. 2008b). Females from diseased populations appear to have more pouch young than females from non-diseased areas (Lazenby et al. 2018). Precocial breeding provides reproductive compensation to counter severe disease-caused mortality rates but is not sufficient to significantly slow the decline in population growth rates (Lachish et al. 2009). Precocial breeding may result in rapid evolution of traits leading to a species that is more resilient to DFTD (Jones et al. 2008a).

Much of transmission likely occurs during the mating season, when injurious contacts peak in both males and females (Hamede et al. 2008, 2013b). Patterns of biting injuries and subsequent infections suggest that it is the dominant individuals that are responsible for a large proportion of transmission. As devils with fewer bite injuries had a higher incidence of DFTD (DFT1) it suggests that dominant devils are more likely to acquire DFTD (DFT1) than submissive devils. As the initial tumors are more likely to be inside the oral cavity, it is feasible that the dominant individuals are biting into the tumors of diseased devils (Hamede et al. 2013b). This concords with observations that the most reproductively fit devils are those more likely to become infected (Wells et al. 2017). The diseased devils could then transmit DFTD when they are bitten on the face.

Heterogeneity in epidemic patterns has been observed across Tasmania. For example there was a reduced impact at one site in north-western Tasmania, at one site in north-western Tasmania (West Pencil Pine) (Hamede et al. 2012). At this site disease prevalence remained low, there was no population decline and age structure remained unaltered for up to 5 years after the epidemic outbreak (Hamede et al. 2012). Three plausible explanations have been postulated to explain these differences in epidemic outcomes among affected populations: (1) lower host susceptibility to infection, (2) lower intraspecific bite rates, and (3) differences in virulence among tumor genetic sublineages. To date, no differences have been found in MHC copy number variation between infected and healthy individuals (Lane et al. 2012) and bite rates among populations that differ in epidemic outcomes are not significantly different from each other (Hamede et al. 2013a), suggesting that tumor genetic sublineages may have a role in driving transmission dynamics and epidemic outcome. This has been recently documented at West Pencil Pine where a sudden localized replacement of tumor genetic sublineages resulted in a significant increase in DFTD prevalence, sudden population decline, and mortality in adult devils (Hamede et al. 2015). The competition between tumor sublineages and the changes in epidemiological parameters suggests that the tumor sub lineages and susceptibility to infection are under strong selective pressure.

Risk factors and prevention strategies

The Tasmanian State Government provided the first level of protection for the species by estabishing an Insurance Population within Tasmania. This metapopulation now contains over 700 animals housed in 44 institutions throughout Australia, America, Europe, and New Zealand. Fortunately, Tasmanian devils are amenable to captive breeding and the Insurance Population has been a resounding success. However, protection for wild devils is not so easily secured. Removal of DFTD from wild populations was trialled between 2004 and 2010, when a disease suppression program was carried out on the Forestier peninsula in south east Tasmania. Animals with external signs of DFTD (DFT1) were euthanized to determine whether this conferred an advantage to the remaining population. Analysis of results showed that this methodology was not resulting in a significant difference when compared with an unmanaged site (Lachish et al. 2010). Further analysis proposed that without access to a pre-diagnostic test (the ability to determine whether a devil had contracted DFTD before showing any outward signs) total removal would be impossible. However, this methodology would do no more than remove animals that DFTD would remove several months later and therefore culling is not an effective option (Beeton and McCallum 2011).

Methods for securing healthy populations of devils in the wild either involve translocations of healthy devils to islands, or involve isolating populations behind barriers. Both of these methods have been adopted by the STDP in trying to return devils to the wild. In 2012 and 2013 a total of 28 devils were released onto Maria Island, a National Park 4 km off the coast of mainland Tasmania. In the following years the devils settled and reproduced, creating an abundant and healthy population of approximately 100 animals (DPIPWE, unpublished data).

Also, in 2012, all devils were removed from the Forestier peninsula to create an isolated area. Barriers were installed to prevent diseased devils from moving into the newly created isolated area. Following years of monitoring to ensure no devils, or disease, remained on the Forestier peninsular, in 2015/16 a population of 49 healthy Tasmanian devils was reintroduced into this protected area, to live wild while still protected from DFTD (DPIPWE, unpublished data).

The key to a successful conservation outcome for the devil and the Tasmanian ecosystem, however, is to augment diminished wild populations. The aim is to return population sizes to a level of ecological functionality, even in areas of disease presence. Releasing healthy endangered animals into an area with a continuing threat faces its own challenges. To this end, trial augmentations of devils to three sites on mainland Tasmania occurred between 2015 and 2017 to determine the best protocol to ensure the greatest chance of survival of devils released back into the wild. Questions posed during the trial included: do captive or wild devils survive better; how far do devils travel on release; how long does it take devils to settle following release; does release directly into the wild (hard release) or indirectly via large enclosures (soft release) confer a greater survival advantage? Results from the releases are still being analyzed but results suggest that wild born devils survive better than captive born devils, which appear to lose their innate wariness of vehicles (Grueber et al. 2017). Additionally, all devils released as part of these trials were given a trial immunization. To establish efficacy would require enough test subjects, in a real life test scenario. These three trial releases provided an opportunity to test this vaccine.

Vaccination approach to DFTD (DFT1)

The first attempts to induce immune responses were in two devils using irradiated cultured DFTD cells with Montanide (Seppic, France) as an adjuvant. Neither of the devils produced evidence of an immune response (Brown et al. 2011). This study did provide evidence for an NK-like activity mediated in the form of antibody dependent cellular cytotoxicity. Thus providing the initial evidence that the immune response of Tasmanian devils did have the capacity to mount cell mediated cytotoxicity. Later studies using either sonicated or irradiated cells with Montanide, but with the addition of the TLR agonist CpG, resulted in antibody and in some cases in vitro cytotoxic responses in 5/6 devils. One of the devils that had seroconverted was challenged with live DFTD (DFT1) cells and appeared to be protected from DFTD. Unfortunately protection was not conferred following a re-challenge with live DFT1 cells. It appeared that the immunization could reduce, rather than prevent, tumor progression (Kreiss et al. 2015).

Allogenic immune responses against MHC-I, that occur in most transplantation rejections, would normally be expected to prevent the engraftment of DFT1 cells. The downregulation of MHC-I expression appears to make the tumors invisible to the new host’s immune system. Although the DFT1 cells do not express the protein for MHC-I, the genes are present and mRNA is produced (Siddle et al. 2007, 2013). Consequently MHC-I expression is under translational or post-translational control. The discovery that MHC-I can be upregulated on DFT1 cells using recombinant IFN-γ (Siddle et al. 2013) provided a new direction for the vaccine research. Upregulation of MHC using IFN-γ in combination with TLR signaling has formed the foundation of ongoing vaccine research. A number of studies also supported an immune based approach to control of DFTD, including the demonstration that activated devil peripheral blood mononuclear cells could kill DFTD cells in vitro (Brown et al. 2016). Injection of MHC-upregulated DFTD cells adjacent to experimentally transferred DFT1 tumors in previously immunized devils triggered immune rejection in half of cases (Tovar et al. 2017). Functional activity of devil TLR receptors was confirmed (Patchett et al. 2015) and the efficacy of a stabilized poly-I: C (Hiltonol®) in combination with imiquimod (TLR-3 and -7 agonists, respectively) was established (Patchett et al. 2017) forming the basis for an experimental vaccine candidate.

Evidence of immunologically mediated tumor regression has been identified in a small proportion of wild (non-immunized) devils with DFTD (DFT1) (Pye et al. 2016a). Such evidence provides support that effective immune responses can be generated against DFTD (DFT1) cancer cells and that protection against DFTD is feasible. Armed with evidence of natural tumor regression combined, with observations that DFTD (DFT1) cancer cells are immunogenic and that IFN-γ upregulated MHC-I expression, preliminary vaccine trials were undertaken. The candidate vaccine consisting of killed MHC upregulated DFTI cells (by treating the cells in vitro with IFN-γ). The adjuvants included ISCOMATRIX® mixed with stabilized poly I: C (Hiltonol®) and imiquimod. Fifty-two captive bred devils destined for release into sites in mainland Tasmania to bolster local populations were vaccinated. This trial demonstrated a class switched IgG response against DFT1 cells in over 95% of devils, demonstrating specific activation of both B and T lymphocytes (Pye et al. 2018). Some vaccinated devils developed DFTD, and when combined with low recapture rates of vaccinated devils, this limits the ability to make inferences about the protective capacity of these responses. However, leukocyte infiltration in these devil’s tumors are more prominent than those in the incumbent population (S. Pye, unpublished data) suggesting immune recognition does occur. Additional signals or immune checkpoint blockade for development of an effective response.

Immunotherapies targeting the immune checkpoint molecules CTLA-4, PD-1, and PD-L1 have recently become the most broadly effective cancer treatment available for humans (Larkin et al. 2015; Tawbi et al. 2018), but this immunotherapy approach is largely untested outside of human medicine. Devil-specific anti-PD-1 and anti-PD-L1 monoclonal antibodies have been developed and used to show that DFT1 and DFT2 cells upregulate the inhibitory molecule PD-L1 in response to IFN-γ stimulation (Flies et al. 2016). However, these anti-PD-1 and anti-PD-L1 monoclonal antibodies were produced in mice, and thus repeated immunotherapy treatment of devils with these antibodies would likely induce an anti-mouse IgG response because the mouse IgG would be viewed as a foreign protein in devils. This response could be ameliorated by “devilizing” the monoclonal antibodies. “Devilizing” in its simplest form would entail producing a recombinant protein that replaces the constant regions of the mouse immunoglobulin heavy and light chains with devil heavy and light chains. Checkpoint molecule immunotherapy on its own is unlikely be used on a large scale, but can be incorporated into vaccines and used to better understand the interaction between DFT cells and the devil immune system. Immunosuppressive cytokines have also been identified as a potential immune evasion mechanism (Morris and Belov 2013), and development and testing of devil cytokines can similarly be used to enhance and inform vaccine development efforts.

Numerous hurdles confront the development of an effective vaccine. The evidence is that the immune response can protect in a minor proportion of wild unvaccinated devils and some vaccinated devils. The immediate challenge is to develop a vaccine that produces a strong response, ideally following a single injection. This would provide a pathway to vaccinate wild devils, in addition to captive devils earmarked for release into the wilderness. The capacity to monitor vaccinated and released devils to test efficacy is also an obstacle to evaluate the efficacy.

What does the future hold concerning DFTD incidence?

In the 20 years since DFTD emerged in Tasmania, there have been no population extinctions but evidence toward the evolution of an endemic disease, coexisting with its host, the Tasmanian devil. DFTD (DFT1, 2, n) is now part of the Tasmanian devil’s ecosystem. Devils and DFTD will evolve and while there are incidences of tumor regression, it is not known if this is an evolutionary response or the result of the interplay of phenotypic response by DFTD or Tasmanian devils. Evidence for evolution is that devils are showing geographically widespread responses to the cancer epidemic, with allele shifts indicating evolving immune-modulated resistance to DFTD(DFT1) occurring within four to six generations of local disease outbreak. Long term coexistence of devil and tumor is further supported by tumor regressions and associated natural immune responses (Pye et al. 2016a), variants in the genome associated with tumor regressions (Wright et al. 2017), and increased tolerance to infection with devils surviving more than a year after infection (Wells et al. 2017). Comparable genetics and mutations between DFT1 and DFT2 suggest a similar epidemic outcome for DFT2.

The STDP was implemented in 2003 by the Tasmanian State Government in response to the decline of the Tasmanian devil population resulting principally from the threat of DFTD. Over the past 15 years, the State and Commonwealth Governments have supported the STDP to determine what DFTD is, how it is transmitted, the effect it has on wild populations once it has been present for a period of time, and importantly, provided the funds to create an insurance population of captive devils to protect against extinction of the species. While our knowledge has informed us that extinction in the wild is unlikely, the overarching goal the STDP has always been to maintain an enduring and sustainable population of devils in the wild. The Wild Devil Recovery trials completed between 2015 and 2017 and the creation of source populations on Maria Island and the Forestier peninsula has set a solid foundation toward this goal.

Devils are susceptible to roadkill, especially in DFTD affected areas where roads are within devil habitat. The loss of a couple of breeding females in a subpopulation could exacerbate devil declines. Consequently, the Tasmanian Government has introduced a roadkill project to mitigate the threat on Tasmanian roads to all Tasmanian wildlife, not just its endangered species. This plan for the future of the devil keeps the devil in the Tasmanian environment, while biding time for a vaccine that provides some protection to the devils released into the wild, and/or possible resistance evolving in the devil or changes to the properties of the cancer cells. Boosting wild population numbers through augmentation can help ameliorate this threat and enhance genetic diversity to provide more devils in the environment to perform their ecological function.

Conclusion

It is now two decades since the first case of DFTD(DFT1). The first decade culminated in the 2006 publication that DFTD(DFT1) is a transmissible cancer. The multidisciplinary approach throughout the second decade has produced considerable progress in immunological and ecological aspects. The lack of any population extinctions and the evidence for rapid evolution toward resistance and disease suggests that DFTD(DFT1) will become an endemic disease. As evidence for evolution has occurred within four to six generations of local disease outbreak it is unlikely that devils will become extinct. However, the conservation outcome for the devil is still uncertain. There remains a need to control increasing incidence and subsequent threats to current Tasmanian devil populations. Efforts toward this aim will incorporate effective vaccination programs. This is based on the evidence that devils, exposed to DFTD in the wild, or as part of an immunization protocol, can mount an immune response to DFTD(DFT1). A vaccination protocol would incorporate DFTD cells, induced to express MHC-I, and adjuvants to provide strong innate immune activation as this has produced immune response in most devils. But a rollout of any vaccination would require evidence of protection against DFTD. Other approaches to protect the devil population include early detection techniques, optimization of disease free breeding program, developing management zones, standards and development of efficient primary and secondary prevention strategies, and ongoing interdisciplinary collaborations.

Funding

Australian Research Council Discovery Grants DP13010075, Australian Research Council Linkage Grant LP130100218, Australian Research Council Discovery Early Career Researcher awards DE170101116 and DE180100484, National Science Foundation Grant DEB 1316549, Dr Eric Guiler Tasmanian Devil Research Grants, Tasmanian Government, Australian Government.

References

  1. Beeton N, McCallum H.. 2011. Models predict that culling is not a feasible strategy to prevent extinction of Tasmanian devils from facial tumour disease. J Appl Ecol 48:1315–23. [Google Scholar]
  2. Bode M, Hawkins C, Rout T, Wintle B.. 2009. Efficiently locating conservation boundaries: searching for the Tasmanian devil facial tumour disease front. Biol Conserv 142:1333–9. [Google Scholar]
  3. Bordes F, Morand S.. 2009. Coevolution between multiple helminth infestations and basal immune investment in mammals: cumulative effects of polyparasitism? Parasitol Res 106:33–7. [DOI] [PubMed] [Google Scholar]
  4. Brock PM, Murdock CC, Martin LB. 2014. The history of ecoimmunology and its integration with disease ecology. Integr Comp Biol 54(3):353–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Brown GK, Kreiss A, Lyons AB, Woods GM.. 2011. Natural killer cell mediated cytotoxic responses in the Tasmanian devil. PLoS One 6:e24475.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Brown GK, Tovar C, Cooray AA, Kreiss A, Darby J, Murphy JM, Corcoran LM, Bettiol SS, Lyons AB, Woods GM.. 2016. Mitogen activated Tasmanian devil blood mononuclear cells kill devil facial tumour disease cells. Immunol Cell Biol 94:673–9. [DOI] [PubMed] [Google Scholar]
  7. Bruniche-Olsen A, Jones ME, Burridge CP, Murchison EP, Holland BR, Austin JJ.. 2018. Ancient DNA tracks the mainland extinction and island survival of the Tasmanian devil. J Biogeogr 45:963–76. [Google Scholar]
  8. Canfield PJ, Cunningham AA.. 1993. Disease and mortality in Australasian marsupials held at London-zoo, 1872–1972. J Zool Wildl Med 24:158–67. [Google Scholar]
  9. Canfield PJ, Hemsley S.. 2000. The roles of histology and immunohistology in the investigation of marsupial disease and normal lymphoid tissue. Dev Comp Immunol 24:455–71. [DOI] [PubMed] [Google Scholar]
  10. Cooper N, Kamilar JM, Nunn CL.. 2012. Host longevity and parasite species richness in mammals. PLoS One 7:e42190.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Coutinho HB, Sewell HF, Tighe P, King G, Nogueira JC, Robalinho TI, Coutinho VB, Cavalcanti VM.. 1995. Immunocytochemical study of the ontogeny of the marsupial Didelphis albiventris immune system. J Anat 187(Pt 1):37–46. [PMC free article] [PubMed] [Google Scholar]
  12. Deakin JE, Belov K.. 2012. A comparative genomics approach to understanding transmissible cancer in Tasmanian devils. Annu Rev Genomics Hum Genet 13:207–22. [DOI] [PubMed] [Google Scholar]
  13. Deakin JE, Bender HS, Pearse AM, Rens W, O’Brien PC, Ferguson-Smith MA, Cheng Y, Morris K, Taylor R, Stuart A, et al. 2012. Genomic restructuring in the Tasmanian devil facial tumour: chromosome painting and gene mapping provide clues to evolution of a transmissible tumour. PLoS Genet 8:e1002483.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Fassati A, Mitchison NA.. 2010. Testing the theory of immune selection in cancers that break the rules of transplantation. Cancer Immunol Immunother 59:643–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Flies AS, Blackburn NB, Lyons AB, Hayball JD, Woods GM.. 2017. Comparative analysis of immune checkpoint molecules and their potential role in the transmissible Tasmanian devil facial tumor disease. Front Immunol 8:513.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Flies AS, Lyons AB, Corcoran LM, Papenfuss AT, Murphy JM, Knowles GW, Woods GM, Hayball JD.. 2016. PD-L1 is not constitutively expressed on Tasmanian devil facial tumor cells but is strongly upregulated in response to IFN-gamma and can be expressed in the tumor microenvironment. Front Immunol 7:581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Griner LA. 1979. Neoplasms in Tasmanian devils (Sarcophilus harrisii). J Natl Cancer Inst 62:589–95. [DOI] [PubMed] [Google Scholar]
  18. Grueber CE, Reid-Wainscoat EE, Fox S, Belov K, Shier DM, Hogg CJ, Pemberton D.. 2017. Increasing generations in captivity is associated with increased vulnerability of Tasmanian devils to vehicle strike following release to the wild. Sci Rep 7:2161.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Guiler ER. 1982. Temporal and spatial distribution of the Tasmanian devil, Sarcophilus harrisi (Dasyuridae: Marsupialia). Pap Proc R Soc Tasmania 116:153–163. [Google Scholar]
  20. Hamede R, Jones ME, McCallum H.. 2013a. Biting injuries and transmission of Tasmanian devil facial tumour disease. J Anim Ecol 82:182–90. [DOI] [PubMed] [Google Scholar]
  21. Hamede R, Lachish S, Belov K, Woods G, Kreiss A, Pearse A-M, Lazenby B, Jones M, McCallum H.. 2012. Reduced effect of Tasmanian devil facial tumor disease at the disease front. Conserv Biol 26:124–34. [DOI] [PubMed] [Google Scholar]
  22. Hamede RK, McCallum H, Jones M.. 2013b. Biting injuries and transmission of Tasmanian devil facial tumour disease. J Anim Ecol 82:182–90. [DOI] [PubMed] [Google Scholar]
  23. Hamede RK, McCallum H, Jones ME.. 2008. Seasonal, demographic and density-related patterns of contact between Tasmanian devils (Sarcophilus harrisii): implications for transmission of devil facial tumour disease. Austral Ecol 33:614–22. [Google Scholar]
  24. Hamede RK, Pearse AM, Swift K, Barmuta LA, Murchison EP, Jones ME.. 2015. Transmissible cancer in Tasmanian devils: localized lineage replacement and host population response. Proc Biol Sci 282:1814. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Hanahan D, Weinberg RA.. 2000. The hallmarks of cancer. Cell 100:57–70. [DOI] [PubMed] [Google Scholar]
  26. Hanahan D, Weinberg RA.. 2011. Hallmarks of cancer: the next generation. Cell 144:646–74. [DOI] [PubMed] [Google Scholar]
  27. Hawkins CE, Baars C, Hesterman H, Hocking GJ, Jones ME, Lazenby B, Mann D, Mooney N, Pemberton D, Pyecroft S, et al. 2006. Emerging disease and population decline of an island endemic, the Tasmanian devil Sarcophilus harrisii. Biol Conserv 131:307–24. [Google Scholar]
  28. Hemsley SW, Canfield PJ, Husband AJ.. 1995. Immunohistological staining of lymphoid tissue in four Australian marsupial species using species cross-reactive antibodies. Immunol Cell Biol 73:321–5. [DOI] [PubMed] [Google Scholar]
  29. Hollings T, Jones M, Mooney N, McCallum H.. 2016. Disease-induced decline of an apex predator drives invasive dominated states and threatens biodiversity. Ecology 97:394–405. [DOI] [PubMed] [Google Scholar]
  30. Howson LJ, Morris KM, Kobayashi T, Tovar C, Kreiss A, Papenfuss AT, Corcoran L, Belov K, Woods GM.. 2014. Identification of dendritic cells, B cell and T cell subsets in Tasmanian devil lymphoid tissue; evidence for poor immune cell infiltration into devil facial tumors. Anat Rec (Hoboken) 297:925–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Hui L, Chen Y.. 2015. Tumor microenvironment: sanctuary of the devil. Cancer Lett 368:7–13. [DOI] [PubMed] [Google Scholar]
  32. Ito R, Takahashi T, Ito M.. 2018. Humanized mouse models: application to human diseases. J Cell Physiol 233:3723–8. [DOI] [PubMed] [Google Scholar]
  33. Johnson CN, Wroe S.. 2003. Causes of extinction of vertebrates during the Holocene of mainland Australia: arrival of the dingo, or human impact? Holocene 13:941–8. [Google Scholar]
  34. Jones ME. 2003. Convergence in ecomorphology and guild structure among marsupial and placental carnivores In: Jones ME, Dickman CR, Archer M, editors. Predators with pouches: the biology of carnivorous marsupials, 285–296. Melbourne, Australia: CSIRO Publishing. [Google Scholar]
  35. Jones ME, Barmuta LA.. 2000. Niche differentiation among sympatric Australian dasyurid carnivores. J Mammal 81:434–47. [Google Scholar]
  36. Jones ME, Cockburn A, Hamede R, Hawkins C, Hesterman H, Lachish S, Mann D, McCallum H, Pemberton D.. 2008. Life-history change in disease-ravaged Tasmanian devil populations. Proc Natl Acad Sci U S A 105:10023–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Jones ME, Cockburn A, Hamede R, Hawkins C, Hesterman H, Lachish S, Mann D, McCallum H, Pemberton D.. 2008. Life-history change in disease-ravaged Tasmanian devil populations. Proc Natl Acad Sci U S A 105:10023–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Jones ME, Paetkau D, Geffen E, Moritz C.. 2004. Genetic diversity and population structure of Tasmanian devils, the largest marsupial carnivore. Mol Ecol 13:2197–209. [DOI] [PubMed] [Google Scholar]
  39. Jurd RD. 1994. “Not proper mammals”: immunity in monotremes and marsupials. Comp Immunol Microbiol Infect Dis 17:41–52. [DOI] [PubMed] [Google Scholar]
  40. Kreiss A, Brown GK, Tovar C, Lyons AB, Woods GM.. 2015. Evidence for induction of humoral and cytotoxic immune responses against devil facial tumor disease cells in Tasmanian devils (Sarcophilus harrisii) immunized with killed cell preparations. Vaccine 33:3016–25. [DOI] [PubMed] [Google Scholar]
  41. Kreiss A, Cheng Y, Kimble F, Wells B, Donovan S, Belov K, Woods GM.. 2011a. Allorecognition in the Tasmanian devil (Sarcophilus harrisii), an endangered marsupial species with limited genetic diversity. PLoS One 6:e22402.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Kreiss A, Fox N, Bergfeld J, Quinn SJ, Pyecroft S, Woods GM.. 2008. Assessment of cellular immune responses of healthy and diseased Tasmanian devils (Sarcophilus harrisii). Dev Comp Immunol 32:544–53. [DOI] [PubMed] [Google Scholar]
  43. Kreiss A, Obendorf DL, Hemsley S, Canfield PJ, Woods GM.. 2009a. A histological and immunohistochemical analysis of lymphoid tissues of the Tasmanian devil. Anat Rec (Hoboken) 292:611–20. [DOI] [PubMed] [Google Scholar]
  44. Kreiss A, Tovar C, Obendorf DL, Dun K, Woods GM.. 2011b. A murine xenograft model for a transmissible cancer in Tasmanian devils. Vet Pathol 48:475–81. [DOI] [PubMed] [Google Scholar]
  45. Kreiss A, Wells B, Woods GM.. 2009b. The humoral immune response of the Tasmanian devil (Sarcophilus harrisii) against horse red blood cells. Vet Immunol Immunopathol 130:135–7. [DOI] [PubMed] [Google Scholar]
  46. Lachish S, Jones M, McCallum H.. 2007. The impact of disease on the survival and population growth rate of the Tasmanian devil. J Anim Ecol 76:926–36. [DOI] [PubMed] [Google Scholar]
  47. Lachish S, McCallum H, Jones M.. 2009. Demography, disease and the devil: life-history changes in a disease-affected population of Tasmanian devils (Sarcophilus harrisii). J Anim Ecol 78:427–36. [DOI] [PubMed] [Google Scholar]
  48. Lachish S, McCallum H, Mann D, Pukk CE, Jones ME.. 2010. Evaluation of selective culling of infected individuals to control Tasmanian devil facial tumor disease. Conserv Biol 24:841–51. [DOI] [PubMed] [Google Scholar]
  49. Lambeck K, Chappell J.. 2001. Sea level change through the last glacial cycle. Science 292:679–86. [DOI] [PubMed] [Google Scholar]
  50. Lane A, Cheng Y, Wright B, Hamede R, Levan L, Jones M, Ujvari B, Belov K.. 2012. New insights into the role of MHC diversity in devil facial tumour disease. PLoS One 7:e36955. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Larkin J, Hodi FS, Wolchok JD.. 2015. Combined Nivolumab and Ipilimumab or monotherapy in untreated melanoma. N Engl J Med 373:1270–1. [DOI] [PubMed] [Google Scholar]
  52. Lazenby BT, Tobler MW, Brown WE, Hawkins CE, Hocking GJ, Hume F, Huxtable S, Iles P, Jones ME, Lawrence C, et al. 2018. Density trends and demographic signals uncover the long‐term impact of transmissible cancer in Tasmanian devils. J Appl Ecol 55:1368–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Letnic M, Fillios M, Crowther MS.. 2012. Could direct killing by larger dingoes have caused the extinction of the thylacine from mainland Australia?. PLoS One 7:e34877.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Loh R, Bergfeld J, Hayes D, O’Hara A, Pyecroft S, Raidal S, Sharpe R.. 2006a. The pathology of devil facial tumor disease (DFTD) in Tasmanian devils (Sarcophilus harrisii). Vet Pathol 43:890–5. [DOI] [PubMed] [Google Scholar]
  55. Loh R, Hayes D, Mahjoor A, O’Hara A, Pyecroft S, Raidal S.. 2006b. The immunohistochemical characterization of devil facial tumor disease (DFTD) in the Tasmanian devil (Sarcophilus harrisii). Vet Pathol 43:896–903. [DOI] [PubMed] [Google Scholar]
  56. McCallum H, Tompkins DM, Jones M, Lachish S, Marvanek S, Lazenby B, Hocking G, Wiersma J, Hawkins CE.. 2007. Distribution and impacts of Tasmanian devil facial tumor disease. EcoHealth 4:318–25. [Google Scholar]
  57. Meek PD, Ballard GA, Vernes K, Fleming PJS.. 2015. The history of wildlife camera trapping as a survey tool in Australia. Aust Mammal 37:1–12. [Google Scholar]
  58. Miller W, Hayes VM, Ratan A, Petersen DC, Wittekindt NE, Miller J, Walenz B, Knight J, Qi J, Zhao F, et al. 2011. Genetic diversity and population structure of the endangered marsupial Sarcophilus harrisii (Tasmanian devil). Proc Natl Acad Sci U S A 108:12348–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Morris K, Belov K.. 2013. Does the devil facial tumour produce immunosuppressive cytokines as an immune evasion strategy?. Vet Immunol Immunopathol 153:159–64. [DOI] [PubMed] [Google Scholar]
  60. Murchison EP, Schulz-Trieglaff OB, Ning Z, Alexandrov LB, Bauer MJ, Fu B, Hims M, Ding Z, Ivakhno S, Stewart C, et al. 2012. Genome sequencing and analysis of the Tasmanian devil and its transmissible cancer. Cell 148:780–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Murchison EP, Tovar C, Hsu A, Bender HS, Kheradpour P, Rebbeck CA, Obendorf D, Conlan C, Bahlo M, Blizzard CA, et al. 2010. The Tasmanian devil transcriptome reveals Schwann cell origins of a clonally transmissible cancer. Science 327:84–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Nimmerjahn F, Ravetch JV.. 2007. Antibodies, Fc receptors and cancer. Curr Opin Immunol 19:239–45. [DOI] [PubMed] [Google Scholar]
  63. Nunn CL, Gittleman JL, Antonovics J.. 2003. A comparative study of white blood cell counts and disease risk in carnivores. Proc Biol Sci 270:347–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Nussey DH, Watt KA, Clark A, Pilkington JG, Pemberton JM, Graham AL, McNeilly TN.. 2014. Multivariate immune defences and fitness in the wild: complex but ecologically important associations among plasma antibodies, health and survival. Proc Biol Sci 281:20132931.. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Parsons RS, Atwood J, Guiler ER, Heddle RW.. 1971. Comparative studies on the blood of monotremes and marsupials. I. Haematology. Comp Biochem Physiol B 39:203–8. [DOI] [PubMed] [Google Scholar]
  66. Patchett AL, Latham R, Brettingham-Moore KH, Tovar C, Lyons AB, Woods GM.. 2015. Toll-like receptor signaling is functional in immune cells of the endangered Tasmanian devil. Dev Comp Immunol 53:123–33. [DOI] [PubMed] [Google Scholar]
  67. Patchett AL, Tovar C, Corcoran LM, Lyons AB, Woods GM.. 2017. The toll-like receptor ligands Hiltonol((R)) (polyICLC) and imiquimod effectively activate antigen-specific immune responses in Tasmanian devils (Sarcophilus harrisii). Dev Comp Immunol 76:352–60. [DOI] [PubMed] [Google Scholar]
  68. Pearse AM, Swift K.. 2006. Allograft theory: transmission of devil facial-tumour disease. Nature 439:549.. [DOI] [PubMed] [Google Scholar]
  69. Pearse AM, Swift K, Hodson P, Hua B, McCallum H, Pyecroft S, Taylor R, Eldridge MD, Belov K.. 2012. Evolution in a transmissible cancer: a study of the chromosomal changes in devil facial tumor (DFT) as it spreads through the wild Tasmanian devil population. Cancer Genet 205:101–12. [DOI] [PubMed] [Google Scholar]
  70. Peck S, Corkrey R, Hamede R, Jones M, Canfield P.. 2015. Hematologic and serum biochemical reference intervals for wild Tasmanian devils (Sarcophilus harrisii). Vet Clin Pathol 44:519–29. [DOI] [PubMed] [Google Scholar]
  71. Peck S, Corkrey R, Hamede R, Jones M, Canfield P.. 2016. Hematologic and serum biochemical changes associated with devil facial tumor disease in Tasmanian devils. Vet Clin Pathol 45:417–29. [DOI] [PubMed] [Google Scholar]
  72. Pemberton D. 1990. Social organisation and behaviour of the Tasmanian devil, Sarcophilus harrisii [Hobart]: University of Tasmania. p. 247.
  73. Pinfold TL, Brown GK, Bettiol SS, Woods GM.. 2014. Mouse model of devil facial tumour disease establishes that an effective immune response can be generated against the cancer cells. Front Immunol 5:251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  74. Prowse TA, Johnson CN, Bradshaw CJ, Brook BW.. 2014. An ecological regime shift resulting from disrupted predator–prey interactions in Holocene Australia. Ecology 95:693–702. [DOI] [PubMed] [Google Scholar]
  75. Pye RJ, Hamede R, Siddle HV, Caldwell A, Knowles GW, Swift K, Kreiss A, Jones ME, Lyons AB, Woods GM.. 2016a. Demonstration of immune responses against devil facial tumour disease in wild Tasmanian devils. Biol Lett 12:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Pye RJ, Pemberton D, Tovar C, Tubio JM, Dun KA, Fox S, Darby J, Hayes D, Knowles GW, Kreiss A, et al. 2016b. A second transmissible cancer in Tasmanian devils. Proc Natl Acad Sci U S A 113:374–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Pye R, Patchett A, McLennan E, Thomson R, Carver S, Fox S, Pemberton D, Kreiss A, Baz Morelli A, Silva A, Pearse MJ, Corcoran LM, Belov K, Hogg CJ, Woods GM, Lyons AB, et al. 2018. Immunization Strategies Producing a Humoral IgG Immune Response against Devil Facial Tumor Disease in the Majority of Tasmanian Devils Destined for Wild Release. Front Immunol 9:259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Siddle HV, Kreiss A, Eldridge MD, Noonan E, Clarke CJ, Pyecroft S, Woods GM, Belov K.. 2007. Transmission of a fatal clonal tumor by biting occurs due to depleted MHC diversity in a threatened carnivorous marsupial. Proc Natl Acad Sci U S A 104:16221–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Siddle HV, Kreiss A, Tovar C, Yuen CK, Cheng YY, Belov K, Swift K, Pearse AM, Hamede R, Jones ME, et al. 2013. Reversible epigenetic down-regulation of MHC molecules by devil facial tumour disease illustrates immune escape by a contagious cancer. Proc Natl Acad Sci U S A 110:5103–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Stammnitz MR, Coorens THH, Gori KC, Hayes D, Fu B, Wang J, Martin-Herranz DE, Alexandrov LB, Baez-Ortega A, Barthorpe S, et al. 2018. The origins and vulnerabilities of two transmissible cancers in Tasmanian devils. Cancer Cell 33:607–19.e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Storfer A, Epstein B, Jones M, Micheletti S, Spear SF, Lachish S, Fox S.. 2017. Landscape genetics of the Tasmanian devil: implications for spread of an infectious cancer. Conserv Genet 18:1287–97. [Google Scholar]
  82. Strakova A, Murchison EP.. 2015. The cancer which survived: insights from the genome of an 11000 year-old cancer. Curr Opin Genet Dev 30:49–55. [DOI] [PubMed] [Google Scholar]
  83. Tawbi HA, Forsyth PA, Algazi A, Hamid O, Hodi FS, Moschos SJ, Khushalani NI, Lewis K, Lao CD, Postow MA, et al. 2018. Combined Nivolumab and Ipilimumab in melanoma metastatic to the brain. N Engl J Med 379:722–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Thalmann S, Wise P, Huxtable S.. 2014. Sentinel camera traps monitor the emergence of infectious disease in Tasmanian devils (Sarcophilus harrisii). Collingwood, Victoria, Australia: CSIRO Publishing. [Google Scholar]
  85. Tovar C, Obendorf D, Murchison EP, Papenfuss AT, Kreiss A, Woods GM.. 2011. Tumor-specific diagnostic marker for transmissible facial tumors of Tasmanian devils: immunohistochemistry studies. Vet Pathol 48:1195–203. [DOI] [PubMed] [Google Scholar]
  86. Tovar C, Pye RJ, Kreiss A, Cheng Y, Brown GK, Darby JM, Malley RC, Siddle HVT, Skjodt K, Kaufman J, et al. 2017. Regression of devil facial tumour disease following immunotherapy in immunised Tasmanian devils. Sci Rep 7:43827. [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Wells K, Hamede RK, Kerlin DH, Storfer A, Hohenlohe PA, Jones ME, McCallum HI.. 2017. Infection of the fittest: devil facial tumour disease has greatest effect on individuals with highest reproductive output. Ecol Lett 20:770–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. White LC, Saltre F, Bradshaw CJA, Austin JJ.. 2018. High-quality fossil dates support a synchronous, Late Holocene extinction of devils and thylacines in mainland Australia. Biol Lett 14:20170642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  89. Williams BK, Nichols JD, Conroy MJ.. 2002. Analysis and management of animal populations: modeling, estimation and decision making. San Diego (CA: ): Academic Press. [Google Scholar]
  90. Woods GM, Howson LJ, Brown GK, Tovar C, Kreiss A, Corcoran LM, Lyons AB.. 2015. Immunology of a transmissible cancer spreading among Tasmanian devils. J Immunol 195:23–9. [DOI] [PubMed] [Google Scholar]
  91. Woods GM, Kreiss A, Belov K, Siddle HV, Obendorf DL, Muller HK.. 2007. The immune response of the Tasmanian devil (Sarcophilus harrisii) and devil facial tumour disease. EcoHealth 4:338–45. [Google Scholar]
  92. Wright B, Willet CE, Hamede R, Jones M, Belov K, Wade CM.. 2017. Variants in the host genome may inhibit tumour growth in devil facial tumours: evidence from genome-wide association. Sci Rep 7:423.. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Integrative and Comparative Biology are provided here courtesy of Oxford University Press

RESOURCES