National Academies Press: OpenBook

Animal Models for Assessing Countermeasures to Bioterrorism Agents (2011)

Chapter: 2 Evaluation of Current and Future TMT-Used Animal Models

« Previous: 1 Introduction
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

2

Evaluation of Current and Future TMT-Used Animal Models

This chapter examines how well specific animal models against biothreats of interest to the Transformational Medical Technologies (TMT) reflect various aspects of the human diseases for which medical countermeasures are being developed. As explained in the Introduction, the TMT seeks to identify and develop new or repurposed medical countermeasures that may have broad-spectrum capability, that is, target a number of pathogens with similar mechanisms of disease causation and pathogenesis. This approach is focused on two major groups, hemorrhagic fever viruses and intracellular bacterial pathogens. The Committee on Animal Models for Assessing Countermeasures to Bioterrorism Agents thinks that currently available animal models for these biothreats, while necessary, are imperfect representations of every aspect of human-pathogen interaction especially with regards to their substitution for “adequate and well-controlled efficacy studies in humans” (FDA 2002, p 37989). Given the ethical mandate of the Animal Rule to not harm human participants in clinical trials that “would involve administering a potentially lethal or permanently disabling toxic substance or organism” (ibid.), these models most likely represent the best approach to develop and test countermeasures and the current efforts have performed as well as could be expected given the limitations listed below. These limitations are critical components to be considered when evaluating the utility of an animal model for efficacy studies1 of the known or unknown pathogens of interest to the TMT:

 

  • Lack of sufficient human clinical data (that is, reliable and sophisticated human clinical markers) and knowledge of the natural history2 of these diseases or threats of interest may hinder the successful correlation of the animal models to the infectious diseases of interest. The more scant the human data, the greater the uncertainty of relevance of the animal model.

_________________________

1 The Committee did not consider animal models used for safety evaluation of products developed under the Animal Rule, as “safety evaluation of products is not addressed in this rule” (FDA 2002, p 37989).

2 Natural history refers to the progression of a disease without any intervention.

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

 

  • Both interspecies and intraspecies variability and the constraints imposed by working in biocontainment facilities lead to methodological differences and results that may not be translatable or comparable across different animal models of the same disease. This is particularly relevant to the anticipated clinical experience of human patients.3
  • Experience with product development and clinical trials for some conventional diseases indicate that animal models often are unreliable surrogates for, or predictors of, efficacy and safety.4,5

Historically, animal models have been relied upon to provide preliminary efficacy data for therapeutics against infectious diseases in support and justification of subsequent definitive efficacy studies in human participants to obtain regulatory approval by the Food and Drug Administration (FDA). Because the preclinical data would be evaluated in the context of knowledge from human studies, any deficiencies in data correlation and extrapolation from the animal models to the human condition would presumably be compensated for by the actual data collected during the human studies. Biothreats represent a special problem in that efficacy studies before an actual event are unlikely to take place. In addition, the actual risk of a biothreat attack is difficult to ascertain. These difficulties are even more pronounced in the case of the “unknown-unknowns”.6

Comparing the evaluation process for bioterrorism countermeasures following the preclinical development stage with that for drugs for which human efficacy studies are possible puts in better perspective the regulatory challenges with which the countermeasure development for TMT (or other biodefense) products is beset. Under optimal circumstances, the current process from drug discovery to FDA approval takes an average of 10 to 15 years and costs more than $1 billion (Tamimi and Ellis 2009). According to some estimates the developmental cost of a single drug has soared from $1.1 billion in 1995 to $1.7 billion in 2002, factoring in the costs of failed prospective drugs (Crawford 2004; Mundae and Östör 2010). Those figures apply equally to biopharmaceuticals and small molecules (DiMasi and Grabowski 2007). To date only about 8% of drugs that successfully enter phase 1 studies eventually are granted market approval by the FDA as compared with 14% in the 1980s. The success rate of pharmaceuticals from the first phase 1 study in humans to market is less than 10% (DiMasi et al. 2010).

The main causes of failure in the clinical trial setting are safety problems, which account for about 20% of the attrition rate, and lack of effectiveness, which accounts for about 40% (Kola and Landis 2004; Peck 2007). Inability to predict these failures before human testing or early in clinical trials dramatically escalates costs. In the infectious disease arena, data from the 10 largest pharmaceutical corporations in the period of 1991-2000 showed a success rate of about 15%, while the average success rate for all indications was 11% (Gilbert et al. 2003). Similarly, DiMasi and colleagues (2010) showed a success rate for systemic infectious disease of 15.6% during 1994 and 2003. It is useful to note that from 1981 to 1992 the success rate of anti-infective drugs was 28.1% and that large biopharmaceutical companies appeared to have a higher success rate of 30.2% for all indications (DiMasi 2001). A key

_________________________

3 Lack of data sharing further compounds differences in methods or lack of reproducibility of results across models (see chapter 5 for further discussion).

4 The limitations of animal models for other disease indications (in addition to those encountered in emerging infectious diseases or biothreats research) have been documented in a number of meta-analyses (see Macleod 2011; Perel et al. 2007; Suntharalingam et al. 2006; van der Worp et al. 2010).

5 As discussed in Developing Animal Models for Use in Animal Rule Licensure: The NIAID Approach (Appendix C, p 111-112), developing animal models in biocontainment requires substantial financial and infrastructure investment.

6 As defined in the introduction, the term “unknown-unknown(s)” refers to pathogen(s) that may not be known or knowable because they currently may not exist. Due to the current or future possibility that they may exist, they are considered potential threats (e.g., a novel, genetically engineered, or created pathogen).

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

question is whether medical countermeasures against emerging infectious diseases and other biothreats have a higher likelihood of success in a (theoretical) human trial. Several facts argue against this possibility and support the notion that achieving a success rate close to that of noncountermeasure drug development can only be considered a best-case scenario:

 

  • The pathogenesis of these rare or even unknown infections is mostly unknown and cannot, therefore, guide the development process.
  • The causative pathogens could be optimized to withstand interventions (e.g., via introduced antibiotic resistance).
  • The clinical setting is probably one of mass infection (which may even be caused by more than one infectious agent) and thus is not comparable to randomized clinical trials of hospitalized patients.
  • Most product development occurs with less than average financial support by entities not experienced in full clinical drug development.
  • The restrictions imposed by biocontainment and the strong reliance on nonhuman primates limit the number of animal studies that could be done.

ANIMAL MODELS ARE ANALOGOUS, NOT HOMOLOGOUS SYSTEMS

On a number of occasions the Animal Rule has been misread resulting in the unrealistic expectation that animal efficacy studies accurately and completely reflect the human condition. Indeed, the term “model” implies that it is not intended to completely replicate the human pathophysiology but rather to provide insight into different aspects of the host-pathogen dynamic. In fact, the Animal Rule is based on the notion that there is enough similarity in the response of animals of different species to a pathogen or a group of pathogens to permit a reasoned method to evaluate product efficacy among those different species (humans being the final target). Prior knowledge of the natural history and progression of the human infection shows that the interplay between host and pathogen may or may not mimic what occurs in humans. Animal models are analogous and not homologous and, by their very nature, display a number of limitations both during different stages of the development process and in the design of the experimental protocols that are applied to these models. For the purpose of this report, homology refers to the similarity in evolutionary origin and physiological function. Analogy refers to the quality of resemblance or similarity in function or appearance but not to the similarity in origin or development (Anderson and Tucker 2006).

Although animal models incorporate a variable degree of homology and analogy, the only homologous model for a human is a human (and even among humans genetic differences affect responses and safety for vaccines and therapeutics; He et al. 2011). Most regular drugs and vaccines are tested for both safety and efficacy in clinical trials where the conditions or diseases of concern are endemic in a population, providing the opportunity to use a truly homologous model. Although efficacy data from animals have been used for decades to drive the exploration of new countermeasures to biological agents and toxins, only in the last decade has there been a need to use research data collected exclusively from analogous models (animals belonging to nonhominid taxa) for the same regulatory approval process as data from humans.

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

CONSIDERATIONS FOR ANIMAL MODELS FOR COUNTERMEASURE DEVELOPMENT

Two of the conditions of the Animal Rule that have to be met for the FDA to use evidence of efficacy derived from animal studies are the following:

 

  1. There is a reasonably well-understood pathophysiological mechanism of the pathogenicity of the infectious agent and its prevention or reduction of symptoms by the product.
  2. The effect is demonstrated in more than one animal model (animals belonging to at least two different species) expected to react with a response predictive for humans unless the effect is demonstrated in animals belonging to a single animal species that represent a sufficiently well-characterized animal model for predicting the response in humans (FDA 2002).

These two conditions often provide some of the biggest hurdles in developing an animal model for countermeasure development. For example, the first condition infers that a large amount of human clinical and pathophysiological data is available to compare with the data derived from the animal model. In many cases, there are sparse to no data on some of the biothreat infections because of their rare geographic distribution and infrequent rate of occurrence. Although autopsy data may be available, they provide little information about the natural history of disease and may be influenced during the terminal stages of infection by a variety of epiphenomena, such as the lack of supportive treatment or the presence of secondary systemic failure. Pathogens with tropism for animals of a single species make the fulfillment of the second condition particularly difficult. Variola virus, the causative agent of smallpox, is a prime example of this problem because in nature it infects only humans. Developing working animal models for variola to replicate the natural progression of smallpox is very difficult if not impossible. Furthermore, although in some cases the model may reflect different aspects of the pathophysiology of smallpox, the actual progression of the illness in animals may be quite different from that observed in humans. The rabbit model for pulmonary anthrax is an example of the latter; the difference in progression can create significant problems for protocols related to product development (see further discussion on page 31).

The significance of the majority of pathogens currently viewed as priorities for biodefense research changed over the last ten years in response to the September 11, 2001, events. Despite the changed status, funds for research of these pathogens were minimal, numbers of researchers specializing in this field were low, and overall research progress was slow. Impeding progress even further, a considerable number of these agents are categorized as Risk Group 3 and 4 pathogens for biosafety and security reasons (Select Agents Regulations; 7 CFR Part 331; 9 CFR Part 121; 42 CFR Part 73), therefore requiring biosafety level 3 or 4 (BSL-3 or -4) containment facilities for any research to be conducted in the United States (ibid.). Accordingly, animals can be experimentally infected with these pathogens only in the appropriate animal biosafety level containment facilities (ABSL-3 or -4).

The following review of several pathogens provides a broad representation of the current status of animal models being developed for efficacy testing and highlights specific challenges common among other models in the context of the Animal Rule, as depicted in Table 2-1.

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

TABLE 2-1 Current State of Animal Model Development for Selected Pathogens in the Context of the Animal Rule

Filoviruses Variola virus Francisella tularensis Bacillus anthracis
Research and product discovery Rodent and nonhuman primate models (Falzarano et al. 2011) Surrogate models used with other poxviruses Predominantly murine models Large body of data
Proof of principle Yes in rodent models Yes for surrogate models Historical information from human challenges Large body of data
FDA Animal Rule Applied for Product Transition
1. Well-understood pathophysiology Limited understanding due to lack of human data Limited understanding of humans (Stanford et al. 2007) Strong pathology but basic mechanistic information lacking Toxin-mediated bacteremia
2. Animals of more than one species Mouse, guinea pig, hamster, nonhuman primates, but limited by #1 Specific human tropism of smallpox challenging Mouse, rat, nonhuman primates Rabbit, nonhuman primates
3. Endpoint clearly related to human benefit Survival, but limited by #1 Survival for surrogate models Survival Survival and decreased morbidity
4. Information for effective human dosing Not applicable at this time Yes for specific antibody responses Correlates of protection not well defined Reasonable correlates

FILOVIRUSES

Among viruses, TMTI focuses on those that cause viral hemorrhagic fevers (VHFs), and among those primarily on VHF-causing filoviruses (marburg-, ebola-, and “cuevaviruses”; see Table 2-2 for virus names and abbreviations). All filoviruses, except Reston virus (RESTV) and Lloviu virus (LLOV), are endemic in Central Africa. RESTV is found in the Philippines and LLOV appears to be endemic in Spain. Human filovirus disease outbreaks are rare events, limited in scope, still unpredictable, and usually occur in rural and underdeveloped areas without sophisticated medical or epidemiological infrastructure. Outbreak intervention often occurs weeks or months after index cases7 are reported to local authorities, and Western-style medical treatment is often hindered not only by nonexistent infrastructure and the lack of trained personnel but also by cultural and especially religious, spiritual constraints. Taken together, these obstacles explain the reasons for the current paucity of available human clinical data on diseases caused by filoviruses.

The lack of basic human pathophysiological information raises the disconcerting possibility that current animal systems for filovirus infections could be only crude approximations of the human

_________________________

7 First disease case in an epidemic within a population (NIH 2011).

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

clinical condition rather than truly analogous models. Currently available animal “models” usually rely on the identification of particular animals that, after infection, develop a disease that has some prominent clinical or pathological markers in common with those observed in infected humans rather than on the thorough characterization of host responses that can be compared directly with those of sick humans. Thus, in the case of filoviruses, the dearth of information on the human patient prevents the development of a clinically defendable animal model. Furthermore, additional collection of human clinical data may render these animals ill-suited for the evaluation of pharmaceuticals or vaccines under the premises of the Animal Rule.

TABLE 2-2 Transformational Medical Technologies Viral Pathogen Focus Group: Filovirusesa

New Taxonomy Outdated Taxonomy (Eighth ICTV Report)

Order Mononegavirales

Order Mononegavirales

Family Filoviridae

Family Filoviridae

Genus Marburgvirus

Genus Marburgvirus

Species Marburg marburgvirus

Species Lake Victoria marburgvirus

Virus 1: Marburg virus (MARV)

Virus: Lake Victoria marburgvirus (MARV)

Virus 2: Ravn virus (RAVV)

Genus Ebolavirus

Genus Ebolavirus

Species Taï Forest ebolavirus

Species Côte d’Ivoire ebolavirus [sic]

Virus: Taï Forest virus (TAFV)

Virus: Côte d’Ivoire ebolavirus [sic] (CIEBOV)

Species Reston ebolavirus

Species Reston ebolavirus

Virus: Reston virus (RESTV)

Virus: Reston ebolavirus (REBOV)

Species Sudan ebolavirus

Species Sudan ebolavirus

Virus: Sudan virus (SUDV)

Virus: Sudan ebolavirus (SEBOV)

Species Zaire ebolavirus

Species Zaire ebolavirus

Virus: Ebola virus (EBOV)

Virus: Zaire ebolavirus (ZEBOV)

Species Bundibugyo ebolavirus

 

Virus: Bundibugyo virus (BDBV)

 
   

Genus “Cuevavirus”

 

Species “Lloviu cuevavirus”

 

Virus: Lloviu virus (LLOV)

 

a Taxa not yet approved by the International Committee on Taxonomy of Viruses (ICTV) are in quotation marks.

SOURCE: Kuhn et al. 2010.

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

Filovirus Infection in Humans

The description of the clinical presentation of humans infected with filoviruses is limited. There are at least eight filoviruses, and the diseases caused by them differ substantially in case numbers, case distribution, and case fatality rates. Moreover, there are few reported cases of some of the viruses. For instance, the clinical presentation of the human disease caused by Bundibugyo virus (BDBV) was reported only once (MacNeil et al. 2010). Similarly, the paucity of information on human infection with Taï Forest virus (TAFV) (only one case described thus far and the patient survived) makes it difficult to extrapolate the symptoms and clinical progression of the disease as observed in a single patient to the population at large (Formenty et al. 1999). It remains uncertain whether humans were ever infected with RESTV or LLOV, as neither has to date been isolated from humans. However, the frequent contact of humans with RESTV-infected swine in the Philippines and the possible frequent exposure of tourists to LLOV-infected bats in Spanish caves suggest that, if humans do get infected by these ebolaviruses, the infections might be without clinical consequences (Barrette et al. 2009). Clinical presentation data on Sudan virus (SUDV) infections have yet to be statistically analyzed (Okware et al. 2002; Smith et al. 1978; WHO 1978). To date, the best-characterized filovirus diseases in human patient cohorts are those caused by Marburg virus (MARV), BDBV, and Ebola virus (EBOV), as shown in Tables 2-3, 2-4, and 2-5 (see table references, pages 22-24). It remains to be seen whether these different viruses cause fundamentally different disease pathogenesis.

Symptoms of filovirus disease are unspecific, are easily confused with many other diseases, and lack a pathognomonic marker that allows for the unequivocal diagnosis of filovirus infection. Unfortunately, autopsies of fatally infected humans have only rarely been performed, partly due to cultural constraints and partly due to safety concerns. For instance, of the 1,912 fatal filovirus infections documented between 1967 and 2010, only 31 have been pathologically examined: eight people infected with MARV/ Ravn virus (RAVV) (five in 1967 and one each in 1975, 1980, and 1987; Gear et al. 1975; Gedigk et al. 1968; Geisbert and Jaax 1998; Smith et al. 1982); 21 people infected with EBOV (three in 1976 and 18 in 1995; Murphy 1978; Zaki and Goldsmith 1999); and two people infected with SUDV in 1976 (Dietrich et al. 1978; Ellis et al. 1978). The autopsies mostly addressed gross anatomy, pathology, and standard histology and did not expand into molecular markers. The collection of more detailed clinical data has been attempted multiple times in the past and failed for numerous reasons, including lack of accessibility to patients, knowledge of ongoing outbreaks, or resistance of patients to be evaluated.

Autopsies of MARV/RAVV-infected patients revealed hemorrhagic diathesis into the skin (maculopapular rash), mucous membranes, and soft tissues. The gallbladders appeared normal, spleens were slightly enlarged, and lymph nodes were swollen. Focal necroses in all organs except lungs, skeletal muscles, and bones were typical findings, but inflammatory reactions were absent with the exception of testes and ovaries. MARV/RAVV was detected in macrophages, fibroblasts, hepatocytes, Kupffer cells, adrenal cells, neuroendocrine cells of the adrenal medulla, and alpha and beta pancreatic islet cells (Gear et al. 1975; Gedigk et al. 1968; Geisbert and Jaax 1998; Kuhn 2008; Smith et al. 1982). The autopsy findings in EBOV-infected patients were similar to those described for MARV/RAVV infections (Murphy 1978; Zaki and Goldsmith 1999), whereas findings in the two autopsied SUDV-infected humans remain controversial because of concomitant parasitic (trematode and nematode) infections (Dietrich et al. 1978; Ellis et al. 1978).

Relatively thorough state-of-the-art molecular analyses of filovirus-infected patients are limited to only a few studies for EBOV- and SUDV-infected patients (Baize et al. 1999, 2002; Hutchinson and Rollin 2007; Leroy et al. 2000, 2001, 2011; Rollin et al. 2007; Sanchez et al. 2004; Wauquier et al. 2010 Attempts to identify disease progression markers have shown that EBOV disease survivors mounted an

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

TABLE 2-3 Symptoms of Marburg Virus-Infected Humans

Clinical Symptom Frequency Observed in Survivors (%) Frequency Observed in Fatal Cases (%)
Abdominal pain 59 57
Anorexia 77 72
Arthralgia or myalgia 55 55
Bleeding from puncture sites 0 7
Bleeding from the gums 23 36
Bleeding from any site 59 71
Chest pain 18 4
Conjuctival infection 14 42
Cough 9 5
Diarrhea 59 56
Difficulty breathing 36 58
Epistaxis 18 34
Fever 100 92
Headaches 73 79
Hematemesis 68 76
Hematoma 0 3
Hemoptysis 9 4
Hiccups 18 44
Lumbar pain 5 8
Malaise or fatigue 86 83
Melena 41 58
Nausea and vomiting 77 76
Petechiae 9 7
Sore throat, odynophagia, or dysphagia 43 43

SOURCE: Adapted from Bausch et al. 2006.

early robust antibody (IgG) response directed against the viral nucleoprotein (NP) and matrix protein VP40, followed by clearance of viral antigen and activation of cytotoxic T cells; in fatal cases, no antibody response was observed concomitant with massive activation of monocytes and macrophages and subsequent massive lymphocyte apoptosis. Moreover, the presence of interleukins IL-1β and IL-6 during symptomatic infections could be used as predictor for nonfatal infections, whereas release of IL-10, IL-1RA, and neopterin could be used as predictor for fatal infections (Leroy et al. 2000; Wauquier et al. 2010). In SUDV patients, the interleukin profile was different; survivors had higher concentrations of interferon α (IFN-α) and fatal cases had higher concentrations of IL-6, IL-8, IL-10, and macrophage inflammatory protein 1β (MIP-1β; Hutchinson and Rollin 2007; Rollin et al. 2007; Sanchez et al. 2004).

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

TABLE 2-4 Symptoms of Ebola Virus-Infected Humans

Clinical Symptom Frequency Observed in Survivors (%) Frequency Observed in Fatal Cases (%)
Abdominal pain 68 62
Abortion 5 2
Anorexia 47 43
Anuria 0 7
Arthralgia or myalgia 79 50
Asthenia 95 85
Bleeding from puncture sites 5 8
Bleeding from the gums 0 15
Bloody stools 5 7
Chest pain 5 10
Conjuctival infection 47 42
Convulsions 0 2
Cough 26 7
Diarrhea 84 86
Dysesthesia 5 0
Epistaxis 0 2
Fever 95 93
Headaches 74 52
Hearing loss 11 5
Hematemesis 0 13
Hematoma 0 2
Hematuria 16 7
Hemoptysis 11 0
Hepatomegaly 5 2
Hiccups 5 17
Lumbar pain 26 12
Maculopapular rasha 16 14
Melena 16 8
Nausea and vomiting 68 73
Petechiae 0 8
Sore throat, odynophagia, or dysphagia 58 56
Splenomegaly 5 2
Tachypnea 0 31
Tinnitus 11 1

a variable detection may be attributed to skin color

SOURCE: Adapted from Bwaka et al. 1999.

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

TABLE 2-5 Symptoms of Bundibugyo Virus-Infected Humans

Clinical Symptom Frequency Observed in Survivors (%) Frequency Observed in Fatal Cases (%)
Abdominal pain 88 93
Anorexia or weight loss 83 80
Arthralgia or myalgia 83 86
Diarrhea 92 87
Difficulty breathing 26 57
Fatigue 96 100
Fever 100 100
Headaches 84 93
Hiccups 17 40
Maculopapular rasha 35 33
Nausea and vomiting 92 87
Sore throat, odynophagia, or dysphagia 43 60

a variable detection may be attributed to skin color

SOURCE: Adapted from MacNeil et al. 2010.

Experimental Filovirus Infection in Animals

The animals currently used in experimental filovirus research are mostly nonhuman primates and rodents (see Table 2-6). The majority of published data from well-established animal models,8 including detailed data on pathogenesis and pathology of disease from African green and rhesus monkeys and cynomolgus macaques, stem from experiments with EBOV or MARV strains (Ebola virus references: Alves et al. 2010; Baskerville et al. 1978, 1985; Bowen et al. 1978; Bray et al. 1998; Connolly et al. 1999; Dadaeva et al. 2006; Geisbert 2003a,b; Jaax et al. 1996; Johnson et al. 1995; Kolesnikova et al. 1997; Pereboeba 1993; Ryabchikova et al. 1993, 1996a, 1998, 1999a, 2004; Vogel et al. 1997; Marburg virus references: Bechtelsheimer et al. 1970; Haas et al. 1968a,b; Korb and Slenczka 1971; Lub et al. 1995; Murphy et al. 1971; Oehlert 1971; Robin et al. 1971; Ryabchikova et al. 1994, 1996b, 1999b; Simpson 1969; Simpson et al. 1968; Warfield et al 2007; Zlotnik 1971; Zlotnik and Simpson 1969). Table 2-7 compares hematological disturbances and mean time to death observed in various nonhuman primate species following EBOV infection. With the possible exception of the hematological responses, nonhuman primates infected with MARV or EBOV roughly reflect the human disease, without significant contradictions between clinical signs and gross pathology.

_________________________

8 “Well-established” refers to animal models that are in use in several BSL-4 facilities and are referred to repeatedly in publications on animal use in filovirus research.

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

TABLE 2-6 Animals Used for the Development of Animal Models for Filovirus Disease

Virus Animal Status of Model
Marburg virus (MARV) Common marmoset (Callithrix jacchus) Model under evaluation, supposedly lethal, unpublished
     
  African green monkey (Chlorocebus aethiops) Well-established lethal model, published
     
  Common squirrel monkey (Saimiri sciureus) Anecdotal lethal “model,” uncharacterized, unpublished
     
  Rhesus monkey (Macaca mulatta) Well-established lethal model, published
     
  Dunkin Hartley and strain 13 guinea pigs Well-established lethal model (requires virus adaptation), published (strain 2 guinea pigs are sometimes also used but their pathology has not been described in detail)
     
  Syrian (golden) hamsters Historical lethal model (requires virus adaptation), basically uncharacterized
     
  BALB/c and SCID BALB/c laboratory mice Recently established model, lethal (requires virus adaptation), published
     
Ravn virus (RAVV) Cynomolgus macaque (Macaca fascicularis) Uncharacterized model, mentioned in publications
     
  Rhesus monkey (Macaca mulatta) Established lethal model, published
     
  BALB/c and SCID-BALB/c laboratory mice Recently established model, lethal (requires virus adaptation), published
     
Bundibugyo virus (BDBV) Cynomolgus macaque (Macaca fascicularis) Model under evaluation, lethal
     
  Rhesus monkey (Macaca mulatta) Model under evaluation, thus far unsuccessful, unpublished
     
Taï Forest virus (TAFV) Cynomolgus macaque (Macaca fascicularis) Established partially lethal model, published
     
  Rhesus monkey (Macaca mulatta) Model under evaluation, no data available
     
Reston virus (RESTV) Cynomolgus macaque (Macaca fascicularis) Well-established model, infrequently lethal, published
     
  Domestic pig (Sus scrofa) Model under evaluation, no data available
     
  African green monkey (Chlorocebus aethiops) Not well-established model, often nonlethal, published
     
Sudan virus (SUDV) African green monkey (Chlorocebus aethiops) Not well-established model, lethal, published
     
  Cynomolgus monkey (macaca fascicularis) Established lethal model, published
     
  Rhesus monkey (Macaca mulatta) Not well-established model, lethal, published
     
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
ICR laboratory mice Anecdotal lethal “model,” uncharacterized, unpublished
     
Ebola virus (EBOV) Common marmoset (Callithrix jacchus) Novel lethal model
     
     
     
  Cynomolgus macaque (Macaca fascicularis) Well-established model, lethal, published
     
  African green monkey (Chlorocebus aethiops) Well-established model, lethal, published
     
  Hamadryas baboon (Papio hamadryas) Well-established model, lethal, published
     
  Rhesus monkey (Macaca mulatta) Well-established model, lethal, published
     
  Domestic pig (Sus scrofa) First experiments published, but lethality unclear
     
  Dunkin Hartley and strain 13 guinea pigs Well-established model, lethal, (requires virus adaptation), published (strain 2 guinea pigs are sometimes also used but their pathology has not been described in detail)
     
  Syrian (golden) hamsters Model under evaluation (requires virus adaptation), supposedly lethal, unpublished
     
  BALB/c, C57BL6, and ICR laboratory mice Well-established model, lethal, (requires virus adaptation), published

SOURCE: Adapted from Kuhn 2008 and references therein.

 

TABLE 2-7 Animal-Specific Hematological Differences in Nonhuman Primate Models of Ebola Virus Disease, Infected with 1-10 LD50

Animal Mean Time to Death Hematological Disturbance
Cynomolgus macaque (Macaca fascicularis) 10-14 days Fibrin depositions
     
African green monkey (Chlorocebus aethiops) 7-8 days Microcirculatory disturbances (capillary stasis, erythrocyte aggregation), organs engorged with blood, no hemorrhage, no fibrin depositions
     
Hamadryas baboon (Papio hamadryas) 9-10 days Erythrocyte diapedesis
     
Rhesus monkey (Macaca mulatta) 7-8 days Fibrin depositions, prominent hemorrhages

SOURCE: Adapted from Kuhn 2008.

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

Table 2-8 compares the clinical signs of various EBOV-infected animal species with those of infected humans and presents a rather well-characterized collection of animal models of filovirus infection.

TABLE 2-8 Comparison of Data from Ebola Virus Animal Models with Data from Humans

Symptom Mice (postvirus adaptation) Guinea Pigs (postvirus adaptation) Nonhuman Primates Humans
Disease duration to death (days) 4-55 6-12 5-10 3-30
Virulence High High High High
Fever No Yes Yes Yes
Peak viremia (plaque-forming unit per milliliter) 7.5 × 107-5.6 × 1011 > 05.2 106-108 106.5
Hemorrhages Variable Rare Dependent on primate type Occasional
Maculopapular rash No No Dependent on primate type Variable (detection often depends on skin color)
Disseminated intravascular coagulation no Data conflicting Yes Yes
Liver enzymes Elevated Elevated Elevated Elevated
Lymphopenia Unknown Yes Yes Yes
Lymphocyte apoptosis Yes Unknown Yes Yes
Thrombocytopenia Yes Yes Yes Yes
Cytokine response Yes Unknown Yes Yes
Nitric oxide level elevation Unknown Unknown Yes Yes

SOURCE: Adapted from Kuhn 2008 and references therein.

Although the time to death for humans extends beyond that of the copresented animal species, it is probably affected by a number of external factors (e.g., whether a patient was hospitalized or received any other care). The extended range of time to death in mice is a characteristic of the EBOV mouse model proposed by Bray and colleagues (1998). In more recent studies, MARV-infected mice die 7-10 days postinfection (Warfield et al. 2009), which is closer to the time of death of human patients. Despite these data, there is currently no consensus in the field on which nonhuman primate model better approximates the course of human infection, in part because of the paucity of cytokine data from the various nonhuman primate models that could be compared with the human data collected in the studies mentioned above. Specifically, biochemical analysis of blood from EBOV-infected cynomolgus macaques and rhesus monkeys revealed increased concentrations of IL -6, whereas IL-2 and IL-10 were

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

rarely detectable (Hensley et al. 2002). A different study using RESTV, rather than a clinically relevant filovirus known to infect humans, revealed a different cytokine activation profile than that shown in human EBOV or SUDV infections (Hutchinson et al. 2001). It is also important to note that RESTV, one of two filoviruses that thus far are thought apathogenic in humans, is virulent in cynomolgus macaques, but not in African green monkeys. The results in cynomolgus macaques raise the question of whether they are indeed valuable heterotypic approximations of humans, given that they should succumb only to EBOV but not to RESTV.

To date, five filoviruses (MARV, RAVV, BDBV, EBOV, and SUDV) are being studied for countermeasure development. Although some of the animal models for the most commonly studied of those viruses, MARV and EBOV, are well established and published, data from animal experiments with the other three have not yet been satisfactorily evaluated for studies of pathogenesis or evaluation of pharmaceuticals or vaccines. Moreover, it is apparent that rodents are not good approximations for human disease for the following reasons: (1) the virus needs to be genetically altered (adapted by serial passage) before it is administered to the animals so that they will succumb; (2) disseminated intravascular coagulation (DIC), which is a prominent symptom of infected humans, does not seem to be a hallmark symptom of their disease; and (3) the typical maculopapular rash is absent.

TULAREMIA

The development of animal models for tularemia is interesting because of the availability of both clinical information regarding direct challenge into humans and data regarding the efficacy of the current investigational new drug (IND) vaccine Live-Vaccine Strain (LVS) to protect human volunteers against direct pulmonary challenges with virulent strain Schu S4 of Francisella tularensis (Hornich and Eigelsbach 1966; McCrumb et al. 1957; Saslaw and Carlisle 1961). Consequently, endpoints (diagnostic and clinical) are available that can be used to judge the worthiness and relevance of a tularemia animal model and possibly refine the experiments in this line of research. On the basis of these data, any comparable animal model would be expected to be (1) very sensitive to infections with Schu S4 (Biovar A) serotypes of Francisella; (2) resistant to infection by high doses of the LVS; and (3) protected from significant morbidity and mortality by prevaccination with LVS.

Nonhuman primates and mice are the most prevalent animal models for primary pulmonary tularemia. Laboratory mice have been extremely useful for dissecting the immune response to F. tularensis and understanding some of the pathophysiology (Coriell et al. 1947; Downs et al. 1949; Ruchman and Foshay 1949). Indeed, the pathology of pyrogranulomae and the primary organ involvement of lung, spleen, and liver are consistent between humans and laboratory mice. However, unlike the human, mice are sensitive to LVS infection, and low doses of LVS do not reproducibly protect these animals from subsequent challenge by Schu S4 (Conlan et al. 2003; Wu et al. 2005); these facts diminish the use of this model for vaccine development. Recently a model based on Fischer 344 rats was shown to be resistant to LVS administration. Further, vaccination with LVS by any route protects these animals against subsequent challenge with relatively high doses of Schu S4 (Wu et al. 2009).

The nonhuman primate model for pulmonary tularemia exhibited similar pathology to that of humans in the course of primary infection, while LVS administration elicited a strong protection against challenge with the Schu S4 strain (Lyons and Wu 2007). If these nonhuman primate models are reproducible, then it is possible that vaccines against F. tularensis could be developed. However, little work has been done to decipher the basic mechanism of protection and immunity in these animals and to determine absolute or relative immune responses as correlates of protection. Because of limited understanding of how the human cellular responses develop antibacterial defenses, it remains hard to

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

develop correlates of protection in humans not only to predict clinical benefits but also to increase confidence in the protection afforded by vaccination. If correlates of protection are known, they may further help advance the research to determine an “effective dose” in humans based on animal experimentation, which is a required element of the Animal Rule.

ANTHRAX9

The challenges facing the production of countermeasures may be highlighted by a discussion of the process applied to the biothreat posed by Bacillus anthracis. B. anthracis has been studied for decades, and the details related to the life cycle of the bacterium are well known (Hugh-Jones and Blackburn 2009); therefore, the development of new products for treatment is expected to be straightforward. The aerosolization of B. anthracis spores is the greatest biothreat risk associated with this pathogen, as pulmonary anthrax is the most lethal form of the disease. Once the spores are inhaled, they are phagocytosed by alveolar macrophages and taken to local lymph nodes where they germinate and disseminate as vegetative bacilli to surrounding tissues via the bloodstream. The timing of this dissemination is unpredictable because it depends on the generation of virulence factors, such as the capsule, which engulfs and protects the bacilli, and the intracellular constitution of the tripartite anthrax toxin. The role of these factors has been well described (Makino et al. 2002; Moayeri and Leppla 2004).

Although the rabbit and many nonhuman primate species are considered the primary animal models for therapeutic product development against B. anthracis, a lot of information has been collected through studies in rodent models. Laboratory mice have always been an attractive model because of (1) the plethora of available tools to dissect the host responses that develop against the aerosol challenge with B. anthracis; (2) their small “footprint” and necessary housing area; and (3) the minimal costs associated with their procurement, care, and use. The murine repertoire of antibodies and T-cell reactivity in response to B. anthracis challenge is generated in a process very similar to that of humans. Across several B. anthracis studies in laboratory mice, the primary difference with the human disease is the dominant virulent factor, which in mice is the capsule (Chand et al. 2009). Encapsulated strains of B. anthracis that do not express toxin remain virulent and lethal in most murine models of anthrax except for the susceptible A/J strain. A/J mice deficient in complement protein C5 die from a toxin-mediated death following infection with low doses of the nonencapsulated Sterne strain (Welkos and Friedlander 1988). In this animal model, where the toxin is the target, the current Anthrax Vaccine-Adsorbed (AVA) vaccine provides robust protection, as do other antitoxin modalities, such as antiserum to recombinant protective antigen (Pitt et al. 2001). On the basis of the limited role for B. anthracis toxins in the infection of laboratory mice, these animals are considered a poor model for human anthrax, whose pathogenesis depends on the virulence of toxin (Heninger et al. 2006).

The rat model is thought to be inadequate because of the high baseline resistance of these animals to infection with B. anthracis spores. Some strains of rats (e.g., Fischer 344), however, display high sensitivity to injected purified toxin and are therefore routinely used to screen antitoxin candidates, such as human monoclonal antibodies (Beall and Dalldorf 1966; Sawada-Hirai et al. 2004). Guinea pigs were used in some seminal studies to describe the trafficking of spores delivered via the lung before dissemination (Ross 1957). Guinea pigs are used in potency assays for the licensed AVA

_________________________

9 The National Institute for Allergy and Infectious Diseases’ efforts to fund research into the standardization of biodefense-related animal models for product development under the Animal Rule deserve credit for advancing the anthrax model in particular and for raising awareness of all models more generally (see Appendix C).

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

vaccine based on the protection observed following challenge with parenterally administered spores (FDA 1973). The understanding of the efficacy of this vaccine dates back to data derived from vaccinated workers in wool processing plants in the 1950s (Brachman et al. 1962). Analysis of these data coupled with the fact that guinea pigs challenged by aerosolized anthrax spores are not reliably protected by the AVA vaccine (Fellows et al. 2001) demonstrate that this animal model is not optimal for vaccine testing and screening. Such a priori knowledge of the expected efficacy of a vaccine in humans is unlikely to be available for the majority of current biothreats.

Rabbits and nonhuman primates, such as rhesus monkeys, are sensitive to pulmonary anthrax and demonstrate many of the pathological findings observed in humans (Vasconcelos et al. 2003; Zaucha et al. 1998). Moreover, the gross lesions seen in the cynomolgus macaque pulmonary anthrax model are similar to those seen in infected humans, including splenomegaly, lymph node enlargement, and hemorrhages in several different organs. Mediastinitis was observed in approximately 30% of the infected animals (Vasconcelos et. al. 2003). As both rabbit and macaque species are well-protected by the AVA vaccine, they have been very useful in the development of prophylactic therapeutics against anthrax (Phipps et al. 2004).

In contrast to the nonhuman primate models, the rabbit provided few, if any, clues to the disease progression. Thus, it has been challenging to develop a reproducible rabbit model for therapeutics to be administered during the dissemination stage of the disease for the following reasons: (1) the rabbit shows very few to no clinical symptoms postinfection, thus the timing for postinfection intervention is not easily discernible; and (2) due to the unpredictable timing of dissemination from the lung into the bloodstream, each animal may need therapeutic intervention at different time points. Because the time from infection to death typically occurs within 48-72 hours, this experiment presents enormous logistical challenges.

Although the rabbits and nonhuman primates appear to be the best models for medical countermeasure development under the regulatory provisions of the Animal Rule, their use poses significant challenges with regard to housing capacity, number of animals needed for statistically meaningful results, and cost of procurement and care. Moreover, societal sensitivities toward the use of nonhuman primates in research pose additional impediments to the continued use of these animals in the development and production of medical countermeasures for emerging infections and biothreats.

LESSONS LEARNED FROM DEVELOPING ANIMAL MODELS FOR THERAPEUTIC PURPOSES AGAINST BIOTHREAT AGENTS

The search for prophylactics and therapeutics against infection of B. anthracis, as summarized in the previous pages, has encountered a number of hurdles, some of which stem from trying to force the animal model to fit the experimental protocol instead of selecting the most appropriate model based on the desired experimental outcome (for an expanded discussion on this issue see Chapter 4, page 56). Furthermore, the process of developing animal models and medical countermeasures has been intimately linked in such a manner that the development and subsequent fitness of the model is determined solely in the context of the countermeasure rather than in a product-neutral fashion. It is important to realize that the value of animal models depends on the context of the scientific question to be investigated.

A number of currently used animal models do not translate well to the human condition. Furthermore, most models are complex and therefore costly to develop. High levels of biocontainment

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

are necessary to safely perform research with these pathogens, and additional restrictions are imposed by their classification as Select Agents.10 These facts coupled with the large numbers of animals necessary for the research and development of countermeasures point to the need to reevaluate the ways these models are developed and used. It would be more beneficial to develop models with a broader application profile that can be used to develop more than one countermeasure.11 Such an approach might address not only the conundrum of over-relying on analogous systems to predict efficacy of products in humans, but may be of significance in an encounter with an “unknown-unknown”.

As previously stated, one of the potential unintended consequences of the Animal Rule is the ambitious expectation that animal efficacy studies predict the human condition. This expectation is daunting for two reasons: (1) there is not enough primary data from humans to which animal data can be compared, and (2) the ability of animal models to reflect the human disease is not absolute. As discussed on page 19, the collection of more detailed clinical data for filovirus infections has been attempted multiple times in the past and failed. At this time, there is no reason to believe that collection of data from filovirus disease outbreaks may improve in the immediate future. Further, while potentially more detailed human data on tularemia and anthrax exists, it is far from comprehensive. The animal models currently available may be the only avenue to accrue some data on pathogenesis, perhaps on correlates of protection, and, through that, on efficacy of pharmaceuticals or vaccines. These circumstances also reflect the TMT’s other concern, namely, the deliberate attack on warfighters with an “unknown-unknown,” that is, an agent for which human clinical data are not available at the time of attack. Therefore, the collection of human clinical data is of utmost importance in order to verify the usefulness and augment the strengths of available models.

The previous sections described the variable results obtained by using animals belonging to different species, a fact also encountered in other fields of biomedical research (e.g., see Craig 2009; Mogil 2009). In addition to factors such as host susceptibility and clinical pathology, the progression of the disease in the different animal species may not resemble that of humans, possibly resulting in failed translational efforts, as evidenced by the increase in attrition rates of products in the later stages of clinical development. In a recent meta-analysis of the potential reasons that animal experiments fail to translate into clinical trials, van der Worp and colleagues (2010) identified recurring themes across animal studies that may prevent them from providing a “correct basis for generalizations to the human condition” as represented in clinical trials (what the authors define as external validity: “the extent to which the results of an animal experiment provide a correct basis for generalizations to the human condition”, p 3; see Table 2-9).

Table 2-9 presents important and common methodological deficiencies, some of which are further discussed in Chapter 5. An additional consideration may be the choice of animals that are young and otherwise healthy, whereas the human patients may have co-morbidities (van der Worp et al. 2010). Addressing some of these issues may be as simple as thoroughly studying the literature. As elaborated further in Chapter 5, however, systematic sharing of data with the wider research community will improve the predictive capacity of animal models. In summary, the more approximation exists between the animals and the conditions under which they are used in efficacy

_________________________

10 “Select Agents” and toxins are agents that the Department of Health and Human Services “considers to have the potential to pose a severe threat to human health. A list of these agents are found in the Select Agents regulation (42 CFR 73).” See http://www.selectagents.gov/FAQ_General.html#sec1q3.

11 The platform technology approach adopted by the TMT fits well with the product-neutral approach. As further discussed in Appendix C, the National Institute of Allergy and Infectious Diseases (NIAID) is similarly focused on product-independent and product-dependent (i.e., product-neutral) models until such time as the product is ready for the final efficacy studies (also known as pivotal studies).

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

studies and the characteristics of the human population for which the countermeasures are intended (including clinical status), the better the chances that the countermeasure would be successful.

TABLE 2-9 Common Causes of Reduced External Validity of Animal Studies

  • Assessment of the effect of a treatment in a homogeneous group of animals versus a heterogeneous group of patients.
  • The use of either male or female animals only, whereas the disease occurs in male and female patients alike.
  • The use of models for inducing a disease or injury with insufficient similarity to the human condition.
  • Delays to start of treatment that are unrealistic in the clinic; the use of doses that are toxic or not tolerated by patients.
  • Differences in outcome measures and the timing of outcome assessment between animal studies and clinical trials.

Source: Adapted from van der Worp et al. 2010.

REFERENCES

Alves DA, Glynn AR, Steele KE, Lackemeyer MG, Garza NL, Buck JG, Mech C, Reed DS. 2010. Aerosol exposure to the Angola strain of Marburg virus causes lethal viral hemorrhagic fever in cynomolgus macaques. Vet Pathol 47(5):831-851.

Anderson J, Tucker K. 2006. Development and validation of animal models. In: Swearengen JR, ed. Biodefense: Research Methodology and Animal Models. Boca Raton: CRC Press. p 41-60.

Baize S, Leroy EM, Georges AJ, Georges-Courbot MC, Capron M, Bedjabaga I, Lansoud-Soukate J, Mavoungou E. 2002. Inflammatory responses in Ebola virus-infected patients. Clin Exp Immunol 128(1):163-168.

Baize S, Leroy EM., Georges-Courbot MC, Capron M, Lansoud-Soukate J, Debré P, Fisher-Hoch SP, McCormick JB, Georges AJ. 1999. Defective humoral responses and extensive intravascular apoptosis are associated with fatal outcome in Ebola virus-infected patients. Nat Med 5(4):423-426.

Barrette RW, Metwally SA, Rowland JM, Xu L, Zaki SR, Nichol ST, Rollin PE, Towner JS, Shieh WJ, Batten B, Sealy TK, Carrillo C, Moran KE, Bracht AJ, Mayr GA, Sirios-Cruz M, Catbagan DP, Lautner EA, Ksiazek TG, White WR, McIntosh MT. 2009. Discovery of swine as a host for the Reston ebolavirus. Science 325(5937):204-206.

Baskerville A, Bowen ET, Platt GS, McArdell LB, Simpson DI. 1978. The pathology of experimental Ebola virus infection in monkeys. J Pathol 125(3):131-138.

Baskerville A, Fisher-Hoch SP, Neild GH, Dowsett AB. 1985. Ultrastructural pathology of experimental Ebola haemorrhagic fever virus infection. J Pathol 147(3):199-209.

Bausch DG, Nichol ST, Muyembe-Tamfum JJ, Borchert M, Rollin PE, Sleurs H, Campbell P, Tshioko FK, Roth C, Colebunders R, Pirard P, Mardel S, Olinda LA, Zeller H, Tshomba A, Kulidri A, Libande ML, Mulangu S, Formenty P, Grein T, Leirs H, Braack L, Ksiazek T, Zaki S, Bowen MD. Smit SB, Leman PA, Burt FJ, Kemp A, Swanepoel R; International Scientific and Technical Committee for Marburg Hemorrhagic Fever Control in the Democratic Republic of the Congo. 2006. Marburg hemorrhagic fever associated with multiple genetic lineages of virus. N Engl J Med 355(9):909-919.

Beall FA, Dalldorf FG. 1966. The pathogenesis of the lethal effect of anthrax toxin in the rat. J Infect Dis 116(3):377-389.

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

Bechtelsheimer H, Korb G, Gedigk P. 1970. The “Marburg-virus” hepatitis. Studies in man and guinea pigs [in German]. Arch A Pathol Pathol Anat 351(4):273-290.

Bowen ET, Platt GS, Simpson DI, McArdell LB, Raymond RT. 1978. Ebola haemorrhagic fever: Experimental infection of monkeys. Trans R Soc Trop Med Hyg 72(2):188-191.

Brachman PS, Gold H, Plotkin SA, Fekety FR, Werrin M, Ingraham NR. 1962. Field evaluation of a human anthrax vaccine. Am J Public Health 52(4):632-645.

Bray M, Davis K, Geisbert T, Schmaljohn C, Huggins J. 1998. A mouse model for evaluation of prophylaxis and therapy of Ebola hemorrhagic fever. J Infect Dis 178(3):651-661.

Bwaka MA, Bonnet MJ, Calain P, Colebunders R, de Roo A, Guimard Y, Katwiki KR, Kibadi K, Kipasa MA, Kuvula KJ, Mapanda BB, Massamba M, Mupapa KD, Muyembe-Tamfum JJ, Ndaberey E, Peters CJ, Rollin PE, van den Enden E. 1999. Ebola hemorrhagic fever in Kikwit, Democratic Republic of the Congo: Clinical observations in 103 patients. J Infect Dis 179(suppl 1): S1-S7.

Carrion R Jr, Ro Y, Hoosien K, Ticer A, Brasky K, de la Garza M, Mansfield K, Patterson JL. 2011. A small nonhuman primate model for filovirus-induced disease. Virology 420(2):117-124.

Chand HS, Drysdale M, Lovchik J, Koehler TM, Lipscomb MF, Lyons CR. 2009. Discriminating virulence mechanisms among Bacillus anthracis strains by using a murine subcutaneous infection model. Infect Immun 77(1):429-435.

Conlan JW, Chen W, Shen H, Webb A, KuoLee R. 2003. Experimental tularemia in mice challenged by aerosol or intradermally with virulent strains of Francisella tularensis: Bacteriologic and histopathologic studies. Microb Pathog 34(5):239-248.

Connolly BM, Steele KE, Davis KJ, Geisbert TW, Kell WM, Jaax NK, Jahrling PB. 1999. Pathogenesis of experimental Ebola virus infection in guinea pigs. J Infect Dis 179(suppl. 1):S203-S217.

Coriell LL, Downs CM, Clapp MP. 1947. Studies on tularemia; immunization of mice. J Immunol 56(3):245-253.

Craig AD. 2009. A rat is not a monkey is not a human: Comment on Mogil (Nature Rev Neurosci 10:283-294 [2009]). Nature Rev Neurosci 10(6):466.

Crawford LM. 2004. Presentation for Banc of America Securities, Healthcare Institutional Investor Conference, July 7, 2004. Available online (http://www.fda.gov/newsEvents/Speeches/ucm053444.htm), accessed August 2011.

Dadaeva AA, Sizikova LP, Subbotina EL, Chepurnov AA. 2006. Hematological and immunological parameters during Ebola virus passages in guinea pigs [in Russian]. Vopr Virusol 51(4):32-37.

Dietrich M, Schumacher HH, Peters D, Knobloch J. 1978. Human pathology of Ebola (Maridi) virus infection in the Sudan. In: Pattyn SR, ed. Ebola Virus Haemorrhagic Fever: Proceedings of an International Colloquium on Ebola Virus Infection and Other Haemorrhagic Fevers, December 6-8, 1977, Antwerp, Belgium. Amsterdam: Elsevier/North-Holland Biomedical Press. p 37-41.

DiMasi JA. 2001. Risks in new drug development: Approved success rates for investigational drugs. Clin Pharmacol Ther 69(5):297-307.

DiMasi JA, Feldman L, Seckler A, Wilson A. 2010. Trends in risks associated with new drug development: Success rates for investigational drugs. Clin Pharmacol Ther 87(3):272-277.

DiMasi JA, Grabowski HG. 2007. The cost of biopharmaceutical R&D: Is biotech different? Manage Decis Econ 28(4-5):469-479.

Downs CM, Buchele L, Edgar EP. 1949. Studies on pathogenesis and immunity in tularemia; the pathogenesis of tularemia in the white rat. J Immunol 63(2):117-133.

Ellis DS, Simpson IH, Francis DP, Knobloch J, Bowen ET, Lolik P, Deng IM. 1978. Ultrastructure of Ebola virus particles in human liver. J Clin Pathol 31(3):201-208.

Falzarano D, Geisbert TW, Feldmann H. 2011. Progress in filovirus vaccine developement: Evaluating the potential for clinical use. Expert Rev Vaccines 10(1):63-77.

FDA [Food and Drug Administration]. 1973. Anthrax Vaccine, Adsorbed. Fed Regist 38 (223):32067-32068 [Established in Title 21 of the Code of Federal Regulations Sections 620.20 - 620.25 for Anthrax Vaccine, Adsorbed (AVA)].

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

FDA. 2002. New Drug and Biological Drug Products; Evidence Needed to Demonstrate Effectiveness of New Drugs When Human Efficacy Studies Are Not Ethical or Feasible. Fed Regist 67(105):37988-37998. Available online (http://www.fda.gov/OHRMS/DOCKETS/98fr/053102a.htm), accessed March 2011.

Fellows PF, Linscott MK, Ivins BE, Pitt ML, Rossi CA, Gibbs PH, Friedlander AM. 2001. Efficacy of a human anthrax vaccine in guinea pigs, rabbits, and rhesus macaques against challenge by Bacillus anthracis isolates of diverse geographical origin. Vaccine 19(23-24):3241-3247.

Formenty P, Hatz C, Le Guenno B, Stoll A, Rogenmoser P, Widmer A. 1999. Human infection due to Ebola virus, subtype Côte d’Ivoire: Clinical and biologic presentation. J Infect Dis 179 (suppl 1):S48-S53.

Gear JS, Cassel GA, Gear AJ, Trappler B, Clausen L, Meyers AM, Kew MC, Bothwell TH, Sher R, Miller GB, Schneider J, Koornhof HJ, Gomperts ED, Isaäcson M, Gear JH. 1975. Outbreak of Marburg virus disease in Johannesburg. Br Med J 4(5995):489-493.

Gedigk P, Bechtelsheimer H, Korb G. 1968. Pathological anatomy of the “Marburg virus”disease (the so-called “Marburg monkey disease”[in German]. Dtsch Med Wochenschr 93(12):590-601, 623, 625.

Geisbert TW. Hensley LE, Larsen T, Young HA, Reed DS, Geisbert JB, Scott DP, Kagan E, Jahrling PB, Davis KJ. 2003a. Pathogenesis of Ebola hemorrhagic fever in cynomolgus macaques: Evidence that dendritic cells are early and sustained targets of infection. Am J Pathol 163(6):2347-2370.

Geisbert TW, Young HA, Jahrling PB, Davis KJ, Larsen T, Kagan E, Hensley LE. 2003b. Pathogenesis of Ebola hemorrhagic fever in primate models: Evidence that hemorrhage is not a direct effect of virus-induced cytolysis of endothelial cells. Am J Pathol 163(6):2371-2382.

Geisbert TW, Jaax NK. 1998. Marburg hemorrhagic fever: Report of a case studied by immunohistochemistry and electron microscopy. Ultrastruct Pathol 22(1):3-17.

Gilbert J, Henske P, Singh A. 2003. Rebuilding big pharma’s business model. In Vivo, the Business and Medicine Report, Windhover Information 21(10). Available online (http://www.bain.com/Images/rebuilding_big_pharma.pdf), accessed July 19, 2011.

Haas R, Maass G, Müller J, Oehlert W. 1968a. Experimental infection of Cercopithecus aethiops with the agent of the Frankfut-Marburg Syndrome (FMS) [in German]. Z Med Mikrobiol Immunol 154(3):210-220.

Haas R, Maass G, Oehlert W. 1968b. Studies on the animal-oriented pathogenicity of a Cercopithecus aethiops transmitted human pathogenic agent [in German]. Med Klin 63(35):1359-1363.

He Y, Hoskins JM, McLeod HL. 2011. Copy number variants in pharmacogenetic genes. Trends Mol Med 17(5):244-251.

Heninger S, Drysdale M, Lovchik J, Hutt J, Lipscomb MF, Koehler TM, Lyons CR. 2006. Toxin-deficient mutants of Bacillus anthracis are lethal in a murine model for pulmonary anthrax. Infect Immun 74(11):6067-6074.

Hensley LE, Young HA, Jahrling PB, Geisbert TW. 2002. Proinflammatory response during Ebola virus infection of primate models: Possible involvement of the tumor necrosis factor receptor superfamily. Immunol Lett 80(3):169-179.

Hornick RB, Eigelsbach HT. 1966. Aerogenic immunization of man with live tularemia vaccine. Bacteriol Rev 30(3):532-538.

Hugh-Jones M, Blackburn J. 2009. The ecology of Bacillus anthracis. Mol Aspects Med 30(6):356-367.

Hutchinson KL, Rollin PE. 2007. Cytokine and chemokine expression in humans infected with Sudan Ebola virus. J Infect Dis 196(suppl 2):S357-S363.

Hutchinson KL, Villinger F, Miranda ME, Ksiazek TG, Peters CJ, Rollin PE. 2001. Multiplex analysis of cytokines in the blood of cynomolgus macaques naturally infected with Ebola virus (Reston serotype). J Med Virol 65(3):561-566.

Jaax NK, Davis KJ, Geisbert TW, Vogel P, Jaax GP, Topper M, Jahrling PB. 1996. Lethal experimental infection of rhesus monkeys with Ebola-Zaire (Mayinga) virus by the oral and conjunctival route of exposure. Arch Pathol Lab Med 120(2):140-155.

Johnson E, Jaax N, White J, Jahrling P. 1995. Lethal experimental infection of rhesus monkeys by aerosolized Ebola virus. Int J Exp Pathol 76(4):227-236.

Kola I, Landis J. 2004. Can the pharmaceutical industry reduce attrition rates? Nat Rev Drug Discov 3(8):711-715.

Kolesnikova LV, Ryabchikova E, Rassadkin YuN, Grazhdantseva AA. 1997. Ultrastructural stereological analysis of monkey lungs during experimental Ebola fever. B Exp Biol Med 123(2):178-181.

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

Korb G, Slenczka W. 1971. Histologic findings in livers and spleens of guinea pigs after infection by the Marburg virus. In: Martini GA, Siegert R, eds. Marburg Virus Disease. Berlin: Springer. p 123-124.

Kuhn JH. 2008. Filoviruses. A Compendium of 40 Years of Epidemiological, Clinical, and Laboratory Studies. Archives of Virology Suppl 20. Vienna: Springer.

Kuhn JH, Becker S, Ebihara H, Geisbert TW, Johnson KM, Kawaoka Y, Lipkin WI, Negredo AI, Netesov SV, Nichol ST, Palacios G, Peters CJ, Tenorio A, Volchkov VE, Jahrling PB. 2010. Proposal for a revised taxonomy of the Family Filoviridae: Classification, names of taxa and viruses, and virus abbreviations. Arch Virol 155(12):2083-2103.

Leroy EM, Baize S, Debré P, Lansoud-Soukate J, Mavoungou E. 2001. Early immune responses accompanying human asymptomatic Ebola infections. Clin Exp Immunol 124(3):453-460.

Leroy EM, Baize S, Volchkov VE, Fisher-Hoch SP, Georges-Courbot MC, Lansoud-Soukate J, Capron M, Debré P, McCormick JB, Georges AJ. 2000. Human asymptomatic Ebola infection and strong inflammatory response. Lancet 355(9222):2210-2215.

Leroy EM, Becquart P, Wauquier N, Baize S. 2011. Evidence for Ebola virus superantigen activity. J Virol 85(8):4041-4042.

Lub MY, Sergeyev AN, Pyankova OG, Pyankov OV, Petrishchenko VA, Kotlyarov LA. 1995. Clinical and virological characterization of the disease in guinea pigs aerogenically infected with Marburg virus [in Russian]. Vopr. Virusol. 40(3):119-121.

Lyons CR, Wu TH. 2007. Animal models of Francisella tularensis infection. Ann NY Acad Sci 1105:238-265.

Macleod M. 2011. Why animal research needs to improve. Nature 477(7366):511.

MacNeil A, Farnon EC, Wamala J, Okware S, Cannon DL, Reed Z, Towner JS, Tappero JW, Lutwama J, Downing R, Nichol ST, Ksiazek TG, Rollin PE. 2010. Proportion of deaths and clinical features in Bundibugyo Ebola virus infection, Uganda. Emer Infect Dis 16(12):1969-1972.

Makino S, Watarai M, Cheun HI, Shirahata T, Uchida I. 2002. Effect of the lower molecular capsule released from the cell surface of Bacillus Anthracis on the pathogenesis of anthrax. J Infect Dis 186(2):227-233.

Marsh GA, Haining J, Robinson R, Foord A, Yamada M, Barr JA, Payne J, White J, Yu M, Bingham J, Rollin PE, Nichol ST, Wang LF, Middleton D. 2011. Ebola reston virus infection of pigs: clinical significance and transmission potential. J Infect Dis 204(suppl 3):S804-S809.

McCrumb FR Jr, Snyder MJ, Woodward TE. 1957. Studies on human infection with Pasteurella tularensis; comparison of streptomycin and chloramphenicol in the prophylaxis of clinical disease. Trans Assoc Am Physicians 70:74-79; discussion 79-80.

Moayeri M, Leppla SH. 2004. The roles of anthrax toxin in pathogenesis. Curr Opin Microbiol 7(1):19-24.

Mogil J. 2009. Animal models of pain: Progress and challenges. Nat Rev Neurosci 10(4):283-294.

Mundae MK, Östör AJ. 2010. The long road of biopharmaceutical drug development: From inception to marketing. QJM 103(1):3-7.

Murphy FA. 1978. Pathology of Ebola virus infection. In: Pattyn SR, ed. Ebola Virus Haemorrhagic Fever: Proceedings of an International Colloquium on Ebola Virus Infection and Other Haemorrhagic Fevers, December 6-8, 1977, Antwerp, Belgium. Amsterdam: Elsevier. p 43-59.

Murphy FA, Simpson DI, Whitfield SG, Zlotnik I, Carter GB. 1971. Marburg virus infection in monkeys: Ultrastructural studies. Lab Invest 24(4):279-291.

NIH [National Institutes of Health]. 2011. Emerging and Re-Emerging Infectious Diseases: Glossary. Curriculum Supplement Series-Grades 9-12. Office of Science Education, NIH. Available online (http://science.education.nih.gov/supplements/nih1/diseases/other/glossary/act1-gloss3.htm#i), accessed March 2011.

Okware SI, Omaswa FG, Zaramba S, Opio A, Lutwama JJ, Kamugisha J, Rwaguma EB, Kagwa P, Lamunu M. 2002. An outbreak of Ebola in Uganda. Trop Med Int Health 7(12):1068-1075.

Oehlert W. 1971. The morphological picture in livers, spleens, and lymph nodes of monkeys and guinea pigs after infection with the “Vervet agent”. In: Martini GA, Siegert R, eds. Marburg Virus Disease. Berlin: Springer. p 144-156.

Peck RW. 2007. Driving earlier clinical attrition: If you want to find the needle burn down the haystack. Considerations for biomarker development. Drug Discov Today 12(7-8):289-294.

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

Pereboeva LA, Tkachev VK, Kolesnikova LV, Krendeleva L, Ryabchikova E, Smolina MP. 1993. The ultrastructural changes in guinea pig organs during the serial passage of the Ebola virus [in Russian]. Vopr Virusol 38(4):179-182.

Perel P, Roberts I, Sena E, Wheble P, Briscoe C, Sandercock P, Macleod M, Mignini LE, Jayaram P, Khan KS. 2007. Comparison of treatment effects between animal experiments and clinical trials: systematic review. BMJ 334(7586):197-202.

Phipps AJ, Premanandan C, Barnewall RE, Lairmore MD. 2004. Rabbit and nonhuman primate models of toxin-targeting human anthrax vaccines. Microbiol Mol Biol Rev 68(4):617-629.

Pitt ML, Little SF, Ivins BE, Fellows P, Barth J, Hewetson J, Gibbs P, Dertzbaugh M, Friedlander AM. 2001. In vitro correlate of immunity in a rabbit model of inhalational anthrax. Vaccine 19(32):4768-4773.

Robin Y, Brès P, Camain R. 1971. Passage of Marburg virus in guinea pigs. In: Martini GA, Siegert R, eds. Marburg Virus Disease. Berlin: Springer. p 117-122.

Rollin PE, Bausch DG, Sanchez A. 2007. Blood chemistry measurements and D-Dimer levels associated with fatal and nonfatal outcomes in humans infected with Sudan Ebola virus. J Infect Dis 196(suppl 2):S364-S371.

Ross JM. 1957. The pathogenesis of anthrax following the administration of spores by the respiratory route. J Pathol Bacteriol 73(2):485-494.

Ruchman I, Foshay L. 1949. Immune response in mice after vaccination with Bacterium tularense. J Immunol 61(3):229-234.

Ryabchikova E, Baranova SG, Tkachev VK, Grazhdantseva AA. 1993. The morphological changes in Ebola infection in guinea pigs [in Russian]. Vopr Virusol 38(4):176-179.

Ryabchikova E, Kolesnikova L, Smolina M, Tkachev V, Pereboeva L, Baranova S, Grazhdantseva A, Rassadkin Y. 1996a. Ebola virus infection in guinea pigs: Presumable role of granulomatous inflammation in pathogenesis. Arch Virol 141(5):909-921.

Ryabchikova E, Strelets L, Kolesnikova L, Pyankov O, Sergeev A. 1996b. Respiratory Marburg virus infection in guinea pigs. Arch Virol 141(11):2177-2190.

Ryabchikova E, Kolesnikova LV, Rassadkin IuN. 1998. Microscopic study of species specific features of hemostatic impairment in Ebola virus-infected monkeys [in Russian]. Vestn Ross Akad Med Nauk 3:51-55.

Ryabchikova E, Kolesnikova LV, Luchko SV. 1999a. An analysis of features of pathogenesis in two animal models of Ebola virus infection. J Infect Dis 179(suppl. 1):S199-S202.

Ryabchikova E, Kolesnikova LV, Netesov SV. 1999b. Animal pathology of filoviral infections. Curr Top Microbiol Immunol 235:145-173.

Ryabchikova E, Smolina M, Grajdantseva A, Rassadkin J. 2004. Ebola virus infection in the guinea pig. In: Klenk HD, Feldmann H, eds. Ebola and Marburg Viruses: Molecular and Cellualar Biology. Wymondham, UK: Horizon Bioscience. p 239-254.

Ryabchikova E, Vorontsova LA, Skripchenko AA, Shestopalov AM, Sandakhchiev LS. 1994. Involvement of internal organs of experimental animals infected with Marburg disease virus [in Russian] Bull Exp Biol Med 117(4):430-434.

Sanchez A, Lukwiya M, Bausch D, Mahanty S, Sanchez AJ, Wagoner KD, Rollin PE. 2004. Analysis of human peripheral blood samples from fatal and nonfatal cases of Ebola (Sudan) hemorrhagic fever: Cellular responses, virus load, and nitric oxide levels. J Virol 78(19):10370-10377.

Saslaw S, Carlisle HN. 1961. Studies with tularemia vaccines in volunteers. IV. Brucella agglutinins in vaccinated and nonvaccinated volunteers challenged with Pasteurella tularensis. Am J Med Sci 242:166-172.

Sawada-Hirai RS, Jiang I, Wang F, Sun SM, Nedellec R, Ruther P, Alvarez A, Millis D, Morrow PR, Kang AS. 2004. Human anti-anthrax protective antigen neutralizing monoclonal antibodies derived from donors vaccinated with Anthrax Vaccine Adsorbed. J Immune Based Ther Vaccines 2(1):5.

Simpson DI. 1969. Vervet monkey disease. Transmission to the hamster. Br J Exp Pathol 50(4):389-392.

Simpson DI, Zlotnik I, Rutter DA. 1968. Vervet monkey disease. Experimental infection of guinea pigs and monkeys with the causative agent. Br J Exp Pathol 49(5):458-464.

Smith DH, Francis F, Simpson DIH. 1978. African haemorrhagic fever in the Southern Sudan, 1976: The clinical manifestations. In: Pattyn SR, ed. Ebola Virus Haemorrhagic Fever: Proceedings of an International

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

      Colloquium on Ebola Virus Infection and Other Haemorrhagic Fevers, December 6-8, 1977, Antwerp, Belgium. Amsterdam: Elsevier/North-Holland Biomedical Press. p 21-26.

Smith DH, Johnson BK, Isaacson M, Swanapoel R, Johnson KM, Killey M, Bagshawe A, Siongok T, Keruga KW. 1982. Marburg-virus disease in Kenya. Lancet 1(8276):816-820.

Stanford MM, McFadden G, Karupiah G, Chaudhri G. 2007. Immunopathogenesis of poxvirus infections: Forecasting the impending storm. Immunol Cell Biol 85(2):93-102.

Suntharalingam G, Perry MR, Ward S, Brett SJ, Castello-Cortes A, Brunner MD, Panoskaltsis N. 2006. Cytokine storm in a phase 1 trial of the anti-CD28 monoclonal antibody TGN1412. N Engl J Med 355(10):1018-1028.

Tamimi NA, Ellis P. 2009. Drug development: From concept to marketing! Nephron Clin Pract 113(3):c125-c131.

van der Worp HB, Howells DW, Sena ES, Porritt MJ, Rewell S, O’Collins V, Macleod MR. 2010. Can animal models of diseases reliably inform human studies? PLoS Med 7(3):e1000245.

Vasconcelos D, Barnewall R, Babin M, Hunt R, Estep J, Nielsen C, Carnes R, Carney J. 2003. Pathology of inhalation anthrax in cynomolgus monkeys (Macaca fascicularis). Lab Invest 83(8):1201-1209.

Vogel P, Connolly B, Abplanalp D, Geisbert TW, Kell WM, Jahrling PB, Jaax NK. 1997. Pathology of experimental Ebola-Zaire (Mayinga) virus infection transmitted to guinea-pigs by oral, conjunctival and tonsillar routes. Cell Vis 4(5):298-307.

Warfield KL, Alves DA, Bradfute SB, Reed DK, VanTongeren S, Kalina WV, Olinger GG, Bavari S. 2007. Development of a model for marburgvirus based on severe-combined immunodeficiency mice. Virol J 4:108.

Warfield KL, Bradfute SB, Wells J, Lofts L, Cooper MT, Alves DA, Reed DK, VanTongeren SA, Mech CA, Bavari S. 2009. Development and characetrization of a mouse model for Marburg hemorrhagic fever. J Virol 83(13):6404-6415.

Wauquier N, Becquart P, Padilla C, Baize S, Leroy EM. 2010. Human fatal Zaire Ebola virus infection is associated with an aberrant innate immunity and with massive lymphocyte apoptosis. PLoS Negl Trop Dis 4(10):e837.

Welkos SL, Friedlander AM. 1988. Pathogenesis and genetic control of resistance to the Sterne strain of Bacillus anthracis. Microb Pathog 4(1):53-69.

WHO [World Health Organization]. 1978. Ebola haemorrhagic fever in Sudan, 1976. Report of a WHO/International Study Team. Bull World Health Organ 56(2):247-270.

Wu TH, Hutt JA, Garrison KA, Berliba LS, Zhou Y, Lyons CR. 2005. Intranasal vaccination induces protective immunity against intranasal infection with virulent Francisella tularensis Biovar A. Infect Immun 73(5):2644-2654.

Wu TH, Zsemlye JL, Statom GL, Hutt JA, Schrader RM, Scrymgeour AA, Lyons CR. 2009. Vaccination of Fischer 344 rats against pulmonary infections by Francisella tularensis type A strains. Vaccine 27(34):4684-4693.

Zaki SR, Goldsmith CS. 1999. Pathologic features of Filovirus infections in humans. Curr Top Microbiol Immunol 235:97-116.

Zhang F, Wu R, Qiang X, Zhou M, Wang P. 2009. Antagonism of alpha2A-adrenoceptor: A novel approach to inhibit inflammatory responses in sepsis. J Mol Med (Berl) 88(3):289-296.

Zaucha GM, Pitt LM, Estep J, Ivins BE, Friedlander AM. 1998. The pathology of experimental anthrax in rabbits exposed by inhalation and subcutaneous inoculation. Arch Pathol Lab Med 122(11):982-992.

Zlotnik I. 1971. “Marburg disease”. The pathology of experimentally infected hamsters. In: Martini GA, Siegert R, eds. Marburg Virus Disease. Berlin: Springer. p 129-135.

Zlotnik I, Simpson DI. 1969. The pathology of experimental vervet monkey disease in hamsters. Br J Exp Pathol 50(4):393-399.

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×

This page intentionally left blank.

Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 15
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 16
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 17
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 18
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 19
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 20
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 21
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 22
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 23
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 24
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 25
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 26
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 27
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 28
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 29
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 30
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 31
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 32
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 33
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 34
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 35
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 36
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 37
Suggested Citation:"2 Evaluation of Current and Future TMT-Used Animal Models." National Research Council. 2011. Animal Models for Assessing Countermeasures to Bioterrorism Agents. Washington, DC: The National Academies Press. doi: 10.17226/13233.
×
Page 38
Next: 3 Ethical and Regulatory Challenges in the Development of Countermeasures »
Animal Models for Assessing Countermeasures to Bioterrorism Agents Get This Book
×
Buy Paperback | $46.00 Buy Ebook | $36.99
MyNAP members save 10% online.
Login or Register to save!
Download Free PDF

The Transformational Medical Technologies (TMT) has been a unique component of the U.S. Department of Defense (DoD) medical biodefense efforts since 2006. Its mission is to advance countermeasure research and development in support of the broader goal of the DoD to protect warfighters from emerging infectious diseases and future genetically engineered biological weapons. The TMT, using advanced science and technology approaches, focused on the development of roadspectrum countermeasures that target common host and pathogen pathways or enhance the host's immune response. Many of these pathogens are lethal or cause such debilitating diseases in humans that it is ethically inappropriate to test the efficacy of these countermeasures in human volunteers. In lieu of human participants, these products may be tested in animals and approved for human use under the provisions of the Food and Drug Administration (FDA)'s 2002 Animal Rule. The reliance on animal models for the development and licensure of medical countermeasures against biothreats is challenging for a number of reasons.

The ad hoc Committee on Animal Models for Assessing Countermeasures to Bioterrorism Agents prepared a consensus report that would address the challenges stemming from developing and testing medical countermeasures against biothreat agents in animal models. Animal Models for Assessing Countermeasures to Bioterrorism Agents evaluates how well the existing TMT-employed or candidate animal models reflect the pathophysiology, clinical picture, and treatment of human disease as related to the agents of interest. The report addresses the process and/or feasibility of developing new animal models for critical biodefense research, placing emphasis on the need for a robust and expeditious validation process in terms of the FDA's Animal Rule. The report also evaluates alternatives to the use of animal models based on the premise of the Three Rs.

  1. ×

    Welcome to OpenBook!

    You're looking at OpenBook, NAP.edu's online reading room since 1999. Based on feedback from you, our users, we've made some improvements that make it easier than ever to read thousands of publications on our website.

    Do you want to take a quick tour of the OpenBook's features?

    No Thanks Take a Tour »
  2. ×

    Show this book's table of contents, where you can jump to any chapter by name.

    « Back Next »
  3. ×

    ...or use these buttons to go back to the previous chapter or skip to the next one.

    « Back Next »
  4. ×

    Jump up to the previous page or down to the next one. Also, you can type in a page number and press Enter to go directly to that page in the book.

    « Back Next »
  5. ×

    Switch between the Original Pages, where you can read the report as it appeared in print, and Text Pages for the web version, where you can highlight and search the text.

    « Back Next »
  6. ×

    To search the entire text of this book, type in your search term here and press Enter.

    « Back Next »
  7. ×

    Share a link to this book page on your preferred social network or via email.

    « Back Next »
  8. ×

    View our suggested citation for this chapter.

    « Back Next »
  9. ×

    Ready to take your reading offline? Click here to buy this book in print or download it as a free PDF, if available.

    « Back Next »
Stay Connected!