Home Medicine Unleashing the power of comparative oncology models in nanomedicine research
Article Publicly Available

Unleashing the power of comparative oncology models in nanomedicine research

  • Carolyn J. Henry

    Carolyn Henry, DVM, MS, is a Professor of Oncology at the University of Missouri (MU), with dual appointments in the College of Veterinary Medicine and the School of Medicine. A graduate of Auburn University and board certified in Veterinary Oncology for over 20 years, Henry has been a faculty member at Washington State University (1993–1997) and MU (1997–present). She is Associate Director of Research for Ellis Fischel Cancer Center (the first academic certified member of the MD Anderson Cancer Network); Associate Dean for Research and Graduate Studies at the College of Veterinary Medicine; and the Provost-appointed One Health Facilitator.

    EMAIL logo
Published/Copyright: March 21, 2015
Become an author with De Gruyter Brill

Abstract

The pathway from discovery of novel candidate drugs, including nanomedicine compounds, to FDA approval is lengthy and may be difficult to navigate. Oftentimes, investigational drugs are appropriately abandoned early in the development pathway due to preclinical failure. Other novel compounds may look quite promising in rodent models and preclinical trials, but prove disappointing when tested in human patients. In fact, only 5% of drugs entering Phase I human cancer clinical trials in the US are ultimately approved. Given the enormous cost, in terms of both financial investment and delay in progress toward improved patient outcome, there is a critical need for a more reliable and efficient process. One solution may be to improve translatability of our preclinical data by including trials in cancer-bearing pet dogs in the drug development pathway.

Introduction

The pace at which medical discoveries have translated into novel therapy options for cancer patients in the US has been frustratingly slow, with few drugs surviving the pathway to FDA approval. In fact, only one of every 5000–10,000 prospective new anticancer agents is eventually approved (1, 2). While many candidate drugs are appropriately withdrawn from the drug development pathway due to failures discovered in preclinical evaluation, many others show promise in rodent models that does not translate to success when applied to human patients. Of drugs that enter Phase I human cancer clinical trials in the US, a mere 5% will ultimately be approved (1, 2). Clearly, there is a critical need for improved drug screening and enhanced efficiency in the process of developmental therapeutics. What if the solution to this problem lay [quite literally] at our feet? Perhaps it does.

The role for companion animal models in cancer nanomedicine research

In their 2013 review article entitled, “Mind the gap: A survey of how cancer drug carriers are susceptible to the gap between research and practice”, Stirland et al. address the issue of the failure of preclinical drug performance in animals to be realized in the human clinical setting, stating, one of the difficulties with cancer in the clinical setting is its inherent heterogeneity and similarity to ‘self’, thus making it difficult to choose a ligand that will be universally expressed by cancer cells yet not expressed by healthy tissue. This is not as much of an issue in preclinical studies that use more homogeneous cancer cell lines and could explain the disparity in results when translating to the clinical setting (3). These limitations of preclinical studies in rodent models are cited repeatedly in the literature, yet the potential for companion animal clinical trials to address this problem is rarely mentioned. Part of this exclusion likely relates to a lack of awareness of spontaneously occurring cancer in pet dogs and its similarity to human cancer. While other companion animals species may also serve as models of spontaneously occurring disesases in people, the domestic dog has been utilized most often in cancer studies due to the prevalence of cancer in this species; thus, this review focuses on canine companion animals.

The pet dog population in the US is estimated at 70 million dogs, with the total number of canine veterinary visits in 2011 reported at 130.4 million (4). Of the disease states that prompt veterinary visits, many share a commonality with human health disorders, including cancer. While most people would not be surprised to learn that an estimated 1.6 million Americans will be diagnosed with cancer this year, a lesser known statistic is that cancer is the leading cause of death in dogs, with approximately 6 million dogs diagnosed with cancer each year in the US (5–7). In fact, nearly one in four dogs will develop cancer during their lifetime (8). Of the 10 most common cancers occurring in men and women, ALL have been reported to develop spontaneously in pet dogs (5). Some canine cancers that have been studied as models of analogous human disease include breast, prostate, bladder, and head and neck cancer, as well as osteosarcoma, lymphoma, and malignant melanoma (9, 10). These naturally-occurring neoplastic diseases in companion animals offer significant advantages over more traditional in vivo models of induced neoplastic disease including similarities in terms of etiology, heterogeneity, biological and metastatic behavior, and clinical response to therapy than does a cancer induced in a laboratory animal with an incompetent immune system (9–12). Many of these advantages are outlined in Table 1.

Table 1:

Advantages of pet dog cancer clinical trials.

General AdvantageUnderlying principles
Cancer develops spontaneously, rather than being artificially inducedImmunocompetent host
Shared environmental factors may underlie etiology
Tumor is heterogeneous and tumor microenvironment is analogous
Diagnostic and staging methods are analagousAdvanced imaging methods such as ultrasound, PET/CT, MRI are available
Staging criteria in veterinary oncology are modeled after WHO criteria
Biomarkers may be similar
Shortened life span compared to peopleShorter time frame for trial outcomes to be reached
Can study drug toxicity and disease behavior over lifetime of patient
More control over confounding variables than with human subjectsLifestyle choices – alcohol, tobacco, drug use
Diet – less variability
Hormonal status – can study disesase in neutered subjects
Ability to study therapy in minimal residual disease settingNatural post-surgical disease behavior can be studied
Monitoring for metastatic disease over lifetime is feasible
Genetic and pedigree information may be availableCanine genome defined
American Kennel Club and other breed registration records retrievable
Trial approval process is simplifiedStandard-of-care not always defined
Much of the animal care is provided by the pet owner at home
Pet owner consents for the trial enrollee
Insurance claims usually not a factor
Biosampling advantagesCan harvest ample variety and quantity of samples from live patient
Repeated biopsy of tumor and normal tissue can provide proof of target
Necropsy is not uncommon
Additional funding opportunities may be availableBreed-specific funding to address breed-related diseases
Animal foundation funding
Veterinary scientific community

The elucidation of the canine genome has provided the promise of selecting canine cancer models that can enhance our understanding of tumor etiology and behavior, as well as aid in the progress of a more targeted, personalized medical approach to cancer care (13). Mapping disease susceptibility by dog breed can provide valuable clues regarding underlying genetic causes. For example, if one identifies a dog breed that is uniquely susceptible to a specific tumor type, whereas another breed is rarely affected, comparing the genomics of the two breeds to the rest of the population may help define the underlying etiology, as well as provide insight as to how to manage or mitigate disease development and progression. The study of comparative oncology facilitates optimal disease model selection for preclinical trials in dogs such that results will be directly applicable to human clinical trial design. If veterinary patients can benefit from the resultant innovation, a win-win dynamic exists that will foster further transdisciplinary discovery and collaboration.

The drug development and approval pathway for nanomedicine is somewhat unique in scope compared to traditional medical therapies. In a systematic review of the current nanomedicine pipeline including both commercialized and investigational nanomedicine products, Etheridge et al. identified 363 potential nanomedicine applications and products under review in 2010 and 2011 (14). Thirty-eight of these were previously approved products that were either being investigated for a new condition or that were serving as the comparator group for a new drug product, whereas the remainder were newly investigated nanomedicines. Of the products under investigation, roughly two-thirds were being evaluated for cancer therapy. These included both “hard” nanostructures such as gold and iron oxide and “soft” nanostructures including micelles, liposomes, emulsions, dendrimers, and other polymeric nanostructures (14).

Some of the properties for which nanoparticles are touted as carrying a potential advantage over traditional systemic cancer therapies are the very properties that may be best studied in cancer-bearing pet animals. Improved drug delivery to the cancer target, decreased toxicity to the patient, and sustained therapeutic drug levels are all potential advantages of nanoparticle therapy. Thus, the ability to assess pharmacokinetics, pharmacodynamics, tolerability, and the long-term effects of nanomedicines is essential and arguably most appropriately done using a syngeneic tumor-bearing animal that can be followed clinically for the natural course of its treated disease (15). This is in contrast to rodent models with artificially-induced or xenografted cancer that experience rapid disease progression, often precluding evaluation of long term effects on metastatic disease progression, drug resistance, and organ toxicity.

Enhanced permeability and retention (EPR) is a phenomenon first described in the 1980s by Hiroshi Maeda whereby macromolecular anticancer therapies selectively extravasate and are retained in solid tumors (16). The EPR effect has been exploited in the clinical development of nanotechnology for cancer therapy. If the EPR hypothesis holds true, nano-sized drug carriers should provide an advantage over traditional therapies in that drug concentration within a tumor should be increased, thus improving efficacy while protecting the rest of the body from the drug and reducing toxicity. This is frequently the case for laboratory animal models used in preclinical studies, but has often failed to materialize in clinical settings (17–22). Perhaps the intermediary step of nanoparticle therapy evaluation in tumor-bearing pet dogs could mitigate the time and expense lost on unsuccessful human clinical trials. Preferential tumor targeting can be confirm via pre- and post-drug administration biosampling methods such as biopsy of normal and tumor tissue, as well as by radiolabeling candidate drugs and studying biodistribution via nuclear scintigraphy. What many researchers do not realize is that the identical imaging modalities available in human medicine are also found in many referral and academic veterinary medical centers.

One unique concern related to the use of nanomedicines is that of persistence in the body and what long-term effects could emerge. Whereas traditional cancer therapies are metabolized and excreted from the body, nanoparticles may persist for months or years. Given that human patients eligible for cancer clinical trials often enroll with very advanced and life-threatening disease, long-term effects of nanoparticle persistence may not be realized in the lifetime of the patients receiving them. In contrast to rodent or laboratory animal studies, companion animal cancer clinical trials permit longer term (life-long) follow-up of treated animals and provide the opportunity to assess for the effects of nanoparticle persistence in tissues. Because standard-of-care therapy is not always established for certain canine diseases, dogs may be enrolled in clinical trials at an earlier disease stage. Financial, as well as altruistic incentives often prompt pet owners to seek clinical trial participation, as does the opportunity to provide cutting-edge therapy to pets that are increasingly viewed in many parts of the world as true family members.

Progress and the path forward

There are a number of real-life examples whereby implementation of veterinary oncology clinical trials in cancer-bearing pets has enabled researchers to gain valuable data to inform clinical trial decisions for subsequent human nanomedicine investigations (23–26). Relevant outcomes of veterinary preclinical trials may include proof-of principle data to support further investigation, PK and PD data to guide dosing decisions, and elucidation of tumor targeting efficacy and drug safety. Access to these resources may be gained via contact through searchable veterinary clinical trials databases and through veterinary specialty organizations engaged in research (27–29). Examples are provided in Table 2. As awareness of these models grows, it is hoped that those conducting medical research will utilize spontaneously-occurring companion animal models of human disease to speed progress and improve efficiency of biomedical research and clinical application of innovations.

Table 2:

Overview of organizations and their websites providing contacts for veterinary cancer specialists and clinical trials.

OrganizationContact website
Veterinary Cooperative Oncology Group (VCOG) and the Veterinary Cancer Society (VCS)www.vetcancertrials.org/
FETCH a Curewww.fetchacure.org/resource-library/clinical-trials/
Morris Animal Foundationwww.morrisanimalfoundation.org/vet-clinics/our-research/
Georgia’s Legacy Canine Cancer Resourcewww.fightcaninecancer.com/clinicaltrials.html
Animal Clinical Investigation (ACI)www.animalci.com/trials
National Cancer Institute, Center for Cancer Researchhttps://ccrod.cancer.gov/confluence/display/CCRCOPWeb/Clinical+Trials
Veterinary Cancer Societyhttp://www.vetcancersociety.org/
American College of Veterinary Internal Medicinewww.ACVIM.org
European College of Veterinary Internal Medicine – Companion Animals (ESVIM-CA)www.ecvim-ca.org/home
European Society of Veterinary Oncology (ESVONC)http://www.esvonc.org/

Corresponding author: Carolyn J. Henry, Department of Veterinary Medicine and Surgery, College of Veterinary Medicine and the Department of Medicine, Division of Hematology and Oncology, School of Medicine; 900 E. Campus Drive, University of Missouri; Columbia, MO 65211, USA, Tel.: +573-882-4690, Fax: +573-884-0705, E-mail:

About the author

Carolyn J. Henry

Carolyn Henry, DVM, MS, is a Professor of Oncology at the University of Missouri (MU), with dual appointments in the College of Veterinary Medicine and the School of Medicine. A graduate of Auburn University and board certified in Veterinary Oncology for over 20 years, Henry has been a faculty member at Washington State University (1993–1997) and MU (1997–present). She is Associate Director of Research for Ellis Fischel Cancer Center (the first academic certified member of the MD Anderson Cancer Network); Associate Dean for Research and Graduate Studies at the College of Veterinary Medicine; and the Provost-appointed One Health Facilitator.

References

1. Gupta SC, Sung B, Prasad S, Webb LJ, Aggarwal BB. Cancer drug discovery by repurposing: teaching new tricks to old dogs. Trends Pharmacol Sci 2013;34:508–17.10.1016/j.tips.2013.06.005Search in Google Scholar PubMed

2. Zamboni WC, Torchilin V, Patri AK, Hrkach J, Stern S, Lee R, et al. Best practices in cancer nanotechnology: perspective from NCI nanotechnology alliance. Clin Cancer Res 2012;18:3229–41.10.1158/1078-0432.CCR-11-2938Search in Google Scholar PubMed PubMed Central

3. Stirland DL, Nichols JW, Miura S, Bae YH. Mind the gap: a survey of how cancer drug carriers are susceptible to the gap between research and practice. J Control Release 2013;172:1045–64.10.1016/j.jconrel.2013.09.026Search in Google Scholar PubMed PubMed Central

4. 2012 US Pet ownership & demographics sourcebook. American Veterinary Medical Association.Search in Google Scholar

5. American Cancer Society. Cancer facts and figures 2014. http://www.cancer.org/acs/groups/content/@research/documents/webcontent/acspc-042151.pdf. Accessed on November 11, 2014.Search in Google Scholar

6. Fleming JM, Creevy KE, Promislow DE. Mortality in North American Dogs from 1984 to 2004: An Investigation into Age-, Size-, and Breed-Related Causes of Death. J Vet Intern Med 2011;25:187–98.10.1111/j.1939-1676.2011.0695.xSearch in Google Scholar PubMed

7. National Cancer Institute Comparative Oncology Program. Available from https://ccrod.cancer.gov/confluence/pages/viewpage.action?pageId=46137977 Accessed on November 11, 2014.Search in Google Scholar

8. Bronson RT. Variation in age at death of dogs of different sexes and breeds. Am J Vet Res 1982;43:2057–9.Search in Google Scholar

9. Rowell JL, McCarthy DO, Alvarez CE. Dog models of naturally occurring cancer. Trends Mol Med 2011;17:380–8.10.1016/j.molmed.2011.02.004Search in Google Scholar PubMed PubMed Central

10. Cekanova M, Rathore K. Animal models and therapeutic molecular targets of cancer: utility and limitations. Drug Des Devel Ther 2014;8:1911–22.10.2147/DDDT.S49584Search in Google Scholar PubMed PubMed Central

11. Henry CJ, Bryan JN. Not lost in translation: how study of diseases in our pets can benefit them and us. MO Med 2013;110:216–9.Search in Google Scholar

12. Ranieri G, Gadaleta CD, Patruno R, Zizzo N, Daidone MG, Hansson MG, et al. A model of study for human cancer: Spontaneous occurring tumors in dogs. Biological features and translation for new anticancer therapies. Crit Rev Oncol Hematol 2013;88:187–97.10.1016/j.critrevonc.2013.03.005Search in Google Scholar PubMed

13. Lindblad-Toh K, Lindblad-Toh K, Wade CM, Mikkelsen TS, Karlsson EK, Jaffe DB, et al. Genome sequence, comparative analysis and haplotype structure of the domestic dog. Nature 2005;438:803–19.10.1038/nature04338Search in Google Scholar PubMed

14. Etheridge ML, Campbell SA, Erdman AG, Haynes CL, Wolf SM, McCullough J. The big picture on nanomedicine: the state of investigational and approved nanomedicine products. Nanomedicine 2013;9:1–14.10.1016/j.nano.2012.05.013Search in Google Scholar PubMed PubMed Central

15. Airley R. Lab reports and cat scans: can veterinary oncology guide our way to new treatments for human cancers? Future Med Chem 2012;4:1391–4.10.4155/fmc.12.81Search in Google Scholar PubMed

16. Maeda H, Fang J, Inutsuka T, Kitamoto Y. Vascular permeability enhancement in solid tumor: various factors, mechanisms involved and its implications. Int Immunopharmacol 2003;3:319–28.10.1016/S1567-5769(02)00271-0Search in Google Scholar

17. Matsumura Y, Kataoka K. Preclinical and clinical studies of anticancer agent-incorporating polymer micelles. Cancer Sci 2009;100:572–9.10.1111/j.1349-7006.2009.01103.xSearch in Google Scholar PubMed

18. Sparreboom A, Scripture CD, Trieu V, Williams PJ, De T, Yang A, et al. Comparative preclinical and clinical pharmacokinetics of a cremophor-free, nanoparticle albumin-bound paclitaxel (ABI-007) and paclitaxel formulated in Cremophor (Taxol). Clin Cancer Res 2005;11:4136–43.10.1158/1078-0432.CCR-04-2291Search in Google Scholar PubMed

19. Duncan R, Gaspar R. Nanomedicine(s) under the microscope. Mol Pharm 2011;8:2101–41.10.1021/mp200394tSearch in Google Scholar PubMed

20. Xu Z, Gu W, Huang J, Sui H, Zhou Z, Yang Y, et al. In vitro and in vivo evaluation of actively targetable nanoparticles for paclitaxel delivery. Int J Pharm 2005;288:361–8.10.1016/j.ijpharm.2004.10.009Search in Google Scholar PubMed

21. Paz-Ares L, Ross H, O’Brien M, Riviere A, Gatzemeier U, Von Pawel J, et al. Phase III trial comparing paclitaxel poliglumex vs docetaxel in the second-line treatment of non-small-cell lung cancer. Br J Cancer 2008;98:1608–13.10.1038/sj.bjc.6604372Search in Google Scholar PubMed PubMed Central

22. O’Brien ME, Wigler N, Inbar M, Rosso R, Grischke E, Santoro A, et al. Reduced cardiotoxicity and comparable efficacy in a phase III trial of pegylated liposomal doxorubicin HCl (CAELYX/Doxil) versus conventional doxorubicin for first-line treatment of metastatic breast cancer. Ann Oncol 2004;15:440–9.10.1093/annonc/mdh097Search in Google Scholar PubMed

23. Hauck ML, LaRue SM, Petros WP, Poulson JM, Yu D, Spasojevic I, et al. Phase I trial of doxorubicin-containing low temperature sensitive liposomes in spontaneous canine tumors. Clin Cancer Res 2006;12:4004–10.10.1158/1078-0432.CCR-06-0226Search in Google Scholar PubMed

24. U’Ren LW, Biller BJ, Elmslie RE, Thamm DH, Dow SW. Evaluation of a novel tumor vaccine in dogs with hemangiosarcoma. J Vet Intern Med 2007;21:113–20.10.1111/j.1939-1676.2007.tb02936.xSearch in Google Scholar

25. Axiak-Bechtel SM, Upendran A, Lattimer JC, Kelsey J, Cutler CS, Selting KA, et al. Gum arabic-coated radioactive gold nanoparticles cause no short-term local or systemic toxicity in the clinically relevant canine model of prostate cancer. Int J Nanomed 2014;9:5001–11.10.2147/IJN.S67333Search in Google Scholar PubMed PubMed Central

26. Kamstock D, Guth A, Elmslie R, Kurzman I, Liggitt D, Coro L, et al. Liposome-DNA complexes infused intravenously inhibit tumor angiogenesis and elicit antitumor activity in dogs with soft tissue sarcoma. Cancer Gene Ther 2006;13:306–17.10.1038/sj.cgt.7700895Search in Google Scholar PubMed

27. Gordon IK, Khanna C. Modeling opportunities in comparative oncology for drug development. ILAR J 2010;51:214–20.10.1093/ilar.51.3.214Search in Google Scholar PubMed PubMed Central

28. Gordon I, Paoloni M, Mazcko C, Khanna C. The comparative oncology trials consortium: using spontaneously occurring cancers in dogs to inform the cancer drug development pathway. PLoS Med 2009;6:e1000161.10.1371/journal.pmed.1000161Search in Google Scholar PubMed PubMed Central

29. Paoloni MC, Tandle A, Mazcko C, Hanna E, Kachala S, Leblanc A, et al. Launching a novel preclinical infrastructure: comparative oncology trials consortium directed therapeutic targeting of TNFalpha to cancer vasculature. PLoS One 2009;4:e4972.10.1371/journal.pone.0004972Search in Google Scholar PubMed PubMed Central

Received: 2014-11-15
Accepted: 2015-2-19
Published Online: 2015-3-21
Published in Print: 2015-4-1

©2015 by De Gruyter

Downloaded on 28.12.2025 from https://www.degruyterbrill.com/document/doi/10.1515/ejnm-2014-0046/html
Scroll to top button