18 results on '"Mazcko C"'
Search Results
2. A Revised Diagnostic Classification of Canine Glioma: Towards Validation of the Canine Glioma Patient as a Naturally Occurring Preclinical Model for Human Glioma
- Author
-
Amy K. LeBlanc, Kevin D. Woolard, Kenneth Aldape, Christina Mazcko, Joanna H. Shih, M. Gerard O'Sullivan, Caterina Giannini, Ingrid Cornax, Craig Horbinski, Andrew D. Miller, Daniel J. Brat, Kara N. Corps, Jennifer W. Koehler, Daniel R. Rissi, Mark R. Gilbert, Chad Frank, Jessica Beck, R. Mark Simpson, Brian F. Porter, C. Ryan Miller, Jason T. Huse, Koehler J.W., Miller A.D., Miller C.R., Porter B., Aldape K., Beck J., Brat D., Cornax I., Corps K., Frank C., Giannini C., Horbinski C., Huse J.T., O'Sullivan M.G., Rissi D.R., Simpson R.M., Woolard K., Shih J.H., Mazcko C., Gilbert M.R., and LeBlanc A.K.
- Subjects
Oncology ,Male ,Human glioma ,Intermediate Filaments ,Canine ,0403 veterinary science ,0302 clinical medicine ,Diagnosis ,Dog ,Comparative ,Cancer ,3'-Cyclic-Nucleotide Phosphodiesterases ,Intermediate Filament ,Brain Neoplasms ,Astrocytoma ,Brain ,04 agricultural and veterinary sciences ,General Medicine ,Glioma ,Diagnostic classification ,Neurology ,Canine Glioma ,030220 oncology & carcinogenesis ,Female ,2',3'-Cyclic-Nucleotide Phosphodiesterases ,medicine.medical_specialty ,040301 veterinary sciences ,Clinical Sciences ,Brain tumor ,Neuropathology ,Pathology and Forensic Medicine ,Veterinarians ,Brain Neoplasm ,Diagnosis, Differential ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Rare Diseases ,Dogs ,Internal medicine ,Physicians ,Glial Fibrillary Acidic Protein ,Genetics ,medicine ,Animals ,neoplasms ,Neurology & Neurosurgery ,Animal ,business.industry ,Neurosciences ,Original Articles ,Oligodendrocyte Transcription Factor 2 ,medicine.disease ,Brain Disorders ,nervous system diseases ,Brain Cancer ,Ki-67 Antigen ,2',3'-Cyclic-Nucleotide Phosphodiesterase ,Physician ,Differential ,Neurology (clinical) ,Oligodendroglioma ,2' ,business - Abstract
The National Cancer Institute-led multidisciplinary Comparative Brain Tumor Consortium (CBTC) convened a glioma pathology board, comprising both veterinarian and physician neuropathologists, and conducted a comprehensive review of 193 cases of canine glioma. The immediate goal was to improve existing glioma classification methods through creation of a histologic atlas of features, thus yielding greater harmonization of phenotypic characterization. The long-term goal was to support future incorporation of clinical outcomes and genomic data into proposed simplified diagnostic schema, so as to further bridge the worlds of veterinary and physician neuropathology and strengthen validity of the dog as a naturally occurring, translationally relevant animal model of human glioma. All cases were morphologically reclassified according to a new schema devised by the entire board, yielding a majority opinion diagnosis of astrocytoma (43, 22.3%), 19 of which were low-grade and 24 high-grade, and oligodendroglioma (134, 69.4%), 35 of which were low-grade and 99 were high-grade. Sixteen cases (8.3%) could not be classified as oligodendroglioma or astrocytoma based on morphology alone and were designated as undefined gliomas. The simplified classification scheme proposed herein provides a tractable means for future addition of molecular data, and also serves to highlight histologic similarities and differences between human and canine glioma.
- Published
- 2018
3. Large Scale Comparative Deconvolution Analysis of the Canine and Human Osteosarcoma Tumor Microenvironment Uncovers Conserved Clinically Relevant Subtypes.
- Author
-
Patkar S, Mannheimer J, Harmon S, Mazcko C, Choyke P, Brown GT, Turkbey B, LeBlanc A, and Beck J
- Abstract
Osteosarcoma is a relatively rare but aggressive cancer of the bones with a shortage of effective biomarkers. Although less common in humans, Osteosarcomas are fairly common in adult pet dogs and have been shown to share many similarities with their human analogs. In this work, we analyze bulk transcriptomic data of 213 primary and 100 metastatic Osteosarcoma samples from 210 pet dogs enrolled in nation-wide clinical trials to uncover three Tumor Microenvironment (TME)-based subtypes: Immune Enriched (IE), Immune Enriched Dense Extra-Cellular Matrix-like (IE-ECM) and Immune Desert (ID) with distinct cell type compositions, oncogenic pathway activity and chromosomal instability. Furthermore, leveraging bulk transcriptomic data of canine primary tumors and their matched metastases from different sites, we characterize how the Osteosarcoma TME evolves from primary to metastatic disease in a standard of care clinical setting and assess its overall impact on clinical outcomes of canines. Most importantly, we find that TME-based subtypes of canine Osteosarcomas are conserved in humans and predictive of progression free survival outcomes of human patients, independently of known prognostic biomarkers such as presence of metastatic disease at diagnosis and percent necrosis following chemotherapy. In summary, these results demonstrate the power of using canines to model the human Osteosarcoma TME and discover novel biomarkers for clinical translation.
- Published
- 2023
- Full Text
- View/download PDF
4. Transcriptional profiling of canine osteosarcoma identifies prognostic gene expression signatures with translational value for humans.
- Author
-
Mannheimer JD, Tawa G, Gerhold D, Braisted J, Sayers CM, McEachron TA, Meltzer P, Mazcko C, Beck JA, and LeBlanc AK
- Subjects
- Humans, Animals, Dogs, Prognosis, Transcriptome, Genomics, Osteosarcoma genetics, Osteosarcoma veterinary, Bone Neoplasms genetics, Bone Neoplasms veterinary
- Abstract
Canine osteosarcoma is increasingly recognized as an informative model for human osteosarcoma. Here we show in one of the largest clinically annotated canine osteosarcoma transcriptional datasets that two previously reported, as well as de novo gene signatures devised through single sample Gene Set Enrichment Analysis (ssGSEA), have prognostic utility in both human and canine patients. Shared molecular pathway alterations are seen in immune cell signaling and activation including TH1 and TH2 signaling, interferon signaling, and inflammatory responses. Virtual cell sorting to estimate immune cell populations within canine and human tumors showed similar trends, predominantly for macrophages and CD8+ T cells. Immunohistochemical staining verified the increased presence of immune cells in tumors exhibiting immune gene enrichment. Collectively these findings further validate naturally occurring osteosarcoma of the pet dog as a translationally relevant patient model for humans and improve our understanding of the immunologic and genomic landscape of the disease in both species., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
5. Deep Domain Adversarial Learning for Species-Agnostic Classification of Histologic Subtypes of Osteosarcoma.
- Author
-
Patkar S, Beck J, Harmon S, Mazcko C, Turkbey B, Choyke P, Brown GT, and LeBlanc A
- Subjects
- Humans, Dogs, Animals, Prognosis, Neural Networks, Computer, Osteosarcoma pathology, Bone Neoplasms pathology
- Abstract
Osteosarcomas (OSs) are aggressive bone tumors with many divergent histologic patterns. During pathology review, OSs are subtyped based on the predominant histologic pattern; however, tumors often demonstrate multiple patterns. This high tumor heterogeneity coupled with scarcity of samples compared with other tumor types render histology-based prognosis of OSs challenging. To combat lower case numbers in humans, dogs with spontaneous OSs have been suggested as a model species. Herein, a convolutional neural network was adversarially trained to classify distinct histologic patterns of OS in humans using mostly canine OS data during training. Adversarial training improved domain adaption of a histologic subtype classifier from canines to humans, achieving an average multiclass F1 score of 0.77 (95% CI, 0.74-0.79) and 0.80 (95% CI, 0.78-0.81) when compared with the ground truth in canines and humans, respectively. Finally, this trained model, when used to characterize the histologic landscape of 306 canine OSs, uncovered distinct clusters with markedly different clinical responses to standard-of-care therapy., (Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
6. Canine and murine models of osteosarcoma.
- Author
-
Beck J, Ren L, Huang S, Berger E, Bardales K, Mannheimer J, Mazcko C, and LeBlanc A
- Subjects
- Animals, Disease Models, Animal, Dogs, Humans, Mice, Bone Neoplasms pathology, Bone Neoplasms veterinary, Dog Diseases therapy, Osteosarcoma pathology, Osteosarcoma veterinary
- Abstract
Osteosarcoma (OS) is the most common malignant bone tumor in children. Despite efforts to develop and implement new therapies, patient outcomes have not measurably improved since the 1980s. Metastasis continues to be the main source of patient mortality, with 30% of cases developing metastatic disease within 5 years of diagnosis. Research models are critical in the advancement of cancer research and include a variety of species. For example, xenograft and patient-derived xenograft (PDX) mouse models provide opportunities to study human tumor cells in vivo while transgenic models have offered significant insight into the molecular mechanisms underlying OS development. A growing recognition of naturally occurring cancers in companion species has led to new insights into how veterinary patients can contribute to studies of cancer biology and drug development. The study of canine cases, including the use of diagnostic tissue archives and clinical trials, offers a potential mechanism to further canine and human cancer research. Advancement in the field of OS research requires continued development and appropriate use of animal models. In this review, animal models of OS are described with a focus on the mouse and tumor-bearing pet dog as parallel and complementary models of human OS.
- Published
- 2022
- Full Text
- View/download PDF
7. Transcriptomic profiling in canines and humans reveals cancer specific gene modules and biological mechanisms common to both species.
- Author
-
Tawa GJ, Braisted J, Gerhold D, Grewal G, Mazcko C, Breen M, Sittampalam G, and LeBlanc AK
- Subjects
- Animals, Biomarkers, Tumor genetics, Bone Neoplasms genetics, Bone Neoplasms veterinary, Computational Biology, Dog Diseases classification, Dogs, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, Lung Neoplasms genetics, Lung Neoplasms veterinary, Lymphoma, B-Cell genetics, Lymphoma, B-Cell veterinary, Lymphoma, T-Cell genetics, Lymphoma, T-Cell veterinary, Melanoma genetics, Melanoma veterinary, Molecular Sequence Annotation, Molecular Targeted Therapy, Neoplasms classification, Oncogenes, Osteosarcoma genetics, Osteosarcoma veterinary, Species Specificity, Translational Research, Biomedical, Dog Diseases genetics, Gene Regulatory Networks, Neoplasms genetics, Neoplasms veterinary
- Abstract
Understanding relationships between spontaneous cancer in companion (pet) canines and humans can facilitate biomarker and drug development in both species. Towards this end we developed an experimental-bioinformatic protocol that analyzes canine transcriptomics data in the context of existing human data to evaluate comparative relevance of canine to human cancer. We used this protocol to characterize five canine cancers: melanoma, osteosarcoma, pulmonary carcinoma, B- and T-cell lymphoma, in 60 dogs. We applied an unsupervised, iterative clustering method that yielded five co-expression modules and found that each cancer exhibited a unique module expression profile. We constructed cancer models based on the co-expression modules and used the models to successfully classify the canine data. These canine-derived models also successfully classified human tumors representing the same cancers, indicating shared cancer biology between canines and humans. Annotation of the module genes identified cancer specific pathways relevant to cells-of-origin and tumor biology. For example, annotations associated with melanin production (PMEL, GPNMB, and BACE2), synthesis of bone material (COL5A2, COL6A3, and COL12A1), synthesis of pulmonary surfactant (CTSH, LPCAT1, and NAPSA), ribosomal proteins (RPL8, RPS7, and RPLP0), and epigenetic regulation (EDEM1, PTK2B, and JAK1) were unique to melanoma, osteosarcoma, pulmonary carcinoma, B- and T-cell lymphoma, respectively. In total, 152 biomarker candidates were selected from highly expressing modules for each cancer type. Many of these biomarker candidates are under-explored as drug discovery targets and warrant further study. The demonstrated transferability of classification models from canines to humans enforces the idea that tumor biology, biomarker targets, and associated therapeutics, discovered in canines, may translate to human medicine., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
8. Comparative Molecular Life History of Spontaneous Canine and Human Gliomas.
- Author
-
Amin SB, Anderson KJ, Boudreau CE, Martinez-Ledesma E, Kocakavuk E, Johnson KC, Barthel FP, Varn FS, Kassab C, Ling X, Kim H, Barter M, Lau CC, Ngan CY, Chapman M, Koehler JW, Long JP, Miller AD, Miller CR, Porter BF, Rissi DR, Mazcko C, LeBlanc AK, Dickinson PJ, Packer RA, Taylor AR, Rossmeisl JH Jr, Woolard KD, Heimberger AB, Levine JM, and Verhaak RGW
- Subjects
- Animals, Dogs, Exome genetics, Humans, Isocitrate Dehydrogenase genetics, Tumor Suppressor Protein p53 genetics, Brain Neoplasms genetics, DNA Methylation genetics, Glioma genetics, Mutation genetics
- Abstract
Sporadic gliomas in companion dogs provide a window on the interaction between tumorigenic mechanisms and host environment. We compared the molecular profiles of canine gliomas with those of human pediatric and adult gliomas to characterize evolutionarily conserved mammalian mutational processes in gliomagenesis. Employing whole-genome, exome, transcriptome, and methylation sequencing of 83 canine gliomas, we found alterations shared between canine and human gliomas such as the receptor tyrosine kinases, TP53 and cell-cycle pathways, and IDH1 R132. Canine gliomas showed high similarity with human pediatric gliomas per robust aneuploidy, mutational rates, relative timing of mutations, and DNA-methylation patterns. Our cross-species comparative genomic analysis provides unique insights into glioma etiology and the chronology of glioma-causing somatic alterations., Competing Interests: Declaration of Interests R.G.W.V. declares equity in Boundless Bio, Inc. A.B.H. receives royalties and milestone payments for licensed intellectual property from Celldex Therapeutics, research grant support from Merck, and is a scientific board member for Caris Life Sciences. The other authors declare no competing interests., (Copyright © 2020 Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
9. NCI Comparative Oncology Program Testing of Non-Camptothecin Indenoisoquinoline Topoisomerase I Inhibitors in Naturally Occurring Canine Lymphoma.
- Author
-
Burton JH, Mazcko C, LeBlanc A, Covey JM, Ji J, Kinders RJ, Parchment RE, Khanna C, Paoloni M, Lana S, Weishaar K, London C, Kisseberth W, Krick E, Vail D, Childress M, Bryan JN, Barber L, Ehrhart EJ, Kent M, Fan T, Kow K, Northup N, Wilson-Robles H, Tomaszewski J, Holleran JL, Muzzio M, Eiseman J, Beumer JH, Doroshow JH, and Pommier Y
- Subjects
- Animals, Antineoplastic Agents chemistry, Bone Marrow drug effects, Clinical Trials as Topic, DNA Topoisomerases, Type I metabolism, Disease Models, Animal, Dogs, Drug Monitoring, Lymphoma metabolism, Lymphoma pathology, Maximum Tolerated Dose, Molecular Targeted Therapy, Topoisomerase I Inhibitors chemistry, Antineoplastic Agents pharmacology, Lymphoma drug therapy, Topoisomerase I Inhibitors pharmacology
- Abstract
Purpose: Only one chemical class of topoisomerase I (TOP1) inhibitors is FDA approved, the camptothecins with irinotecan and topotecan widely used. Because of their limitations (chemical instability, drug efflux-mediated resistance, and diarrhea), novel TOP1 inhibitors are warranted. Indenoisoquinoline non-camptothecin topoisomerase I (TOP1) inhibitors overcome chemical instability and drug resistance that limit camptothecin use. Three indenoisoquinolines, LMP400 (indotecan), LMP776 (indimitecan), and LMP744, were examined in a phase I study for lymphoma-bearing dogs to evaluate differential efficacy, pharmacodynamics, toxicology, and pharmacokinetics., Experimental Design: Eighty-four client-owned dogs with lymphomas were enrolled in dose-escalation cohorts for each indenoisoquinoline, with an expansion phase for LMP744. Efficacy, tolerability, pharmacokinetics, and target engagement were determined., Results: The MTDs were 17.5 mg/m
2 for LMP 776 and 100 mg/m2 for LMP744; bone marrow toxicity was dose-limiting; up to 65 mg/m2 LMP400 was well-tolerated and MTD was not reached. None of the drugs induced notable diarrhea. Sustained tumor accumulation was observed for LMP744; γH2AX induction was demonstrated in tumors 2 and 6 hours after treatment; a decrease in TOP1 protein was observed in most lymphoma samples across all compounds and dose levels, which is consistent with the fact that tumor response was also observed at low doses LMP744. Objective responses were documented for all indenoisoquinolines; efficacy (13/19 dogs) was greatest for LMP744., Conclusions: These results demonstrate proof-of-mechanism for indenoisoquinoline TOP1 inhibitors supporting their further clinical development. They also highlight the value of the NCI Comparative Oncology Program (https://ccr.cancer.gov/Comparative-Oncology-Program) for evaluating novel therapies in immunocompetent pets with cancers., (©2018 American Association for Cancer Research.)- Published
- 2018
- Full Text
- View/download PDF
10. Development of a quantitative pharmacodynamic assay for apoptosis in fixed tumor tissue and its application in distinguishing cytotoxic drug-induced DNA double strand breaks from DNA double strand breaks associated with apoptosis.
- Author
-
Dull AB, Wilsker D, Hollingshead M, Mazcko C, Annunziata CM, LeBlanc AK, Doroshow JH, Kinders RJ, and Parchment RE
- Abstract
DNA double strand breaks (DSBs) induced by cancer therapeutic agents can lead to DNA damage repair or persistent DNA damage, which can induce apoptotic cell death; however, apoptosis also induces DSBs independent of genotoxic insult. γH2AX is an established biomarker for DSBs but cannot distinguish between these mechanisms. Activated cleaved caspase-3 (CC3) promotes apoptosis by enhancing nuclear condensation, DNA fragmentation, and plasma membrane blebbing. Here, we describe an immunofluorescence assay that distinguishes between apoptosis and drug-induced DSBs by measuring coexpression of γH2AX and membrane blebbing-associated CC3 to indicate apoptosis, and γH2AX in the absence of CC3 blebbing to indicate drug-induced DNA damage. These markers were examined in xenograft models following treatment with topotecan, cisplatin, or birinapant. A topotecan regimen conferring tumor regression induced tumor cell DSBs resulting from both apoptosis and direct DNA damage. In contrast, a cisplatin regimen yielding tumor growth delay, but not regression, resulted in tumor cell DSBs due solely to direct DNA damage. MDA-MB-231 xenografts exposed to birinapant, which promotes apoptosis but does not directly induce DSBs, exhibited dose-dependent increases in colocalized γH2AX/CC3 blebbing in tumor cells. Clinical feasibility was established using formalin-fixed, paraffin-embedded biopsies from a canine cancer clinical trial; γH2AX/CC3 colocalization analysis revealed apoptosis induction by two novel indenoisoquinoline topoisomerase I inhibitors, which was consistent with pathologist-assessed apoptosis and reduction of tumor volume. This assay is ready for use in clinical trials to elucidate the mechanism of action of investigational agents and combination regimens intended to inflict DNA damage, apoptotic cell death, or both., Competing Interests: CONFLICTS OF INTEREST The authors declare no potential conflicts of interest.
- Published
- 2018
- Full Text
- View/download PDF
11. Creation of an NCI comparative brain tumor consortium: informing the translation of new knowledge from canine to human brain tumor patients.
- Author
-
LeBlanc AK, Mazcko C, Brown DE, Koehler JW, Miller AD, Miller CR, Bentley RT, Packer RA, Breen M, Boudreau CE, Levine JM, Simpson RM, Halsey C, Kisseberth W, Rossmeisl JH Jr, Dickinson PJ, Fan TM, Corps K, Aldape K, Puduvalli V, Pluhar GE, and Gilbert MR
- Subjects
- Animals, Dogs, Humans, National Cancer Institute (U.S.), United States, Biomedical Research, Brain Neoplasms diagnosis, Brain Neoplasms therapy, Disease Models, Animal
- Abstract
On September 14-15, 2015, a meeting of clinicians and investigators in the fields of veterinary and human neuro-oncology, clinical trials, neuropathology, and drug development was convened at the National Institutes of Health campus in Bethesda, Maryland. This meeting served as the inaugural event launching a new consortium focused on improving the knowledge, development of, and access to naturally occurring canine brain cancer, specifically glioma, as a model for human disease. Within the meeting, a SWOT (strengths, weaknesses, opportunities, and threats) assessment was undertaken to critically evaluate the role that naturally occurring canine brain tumors could have in advancing this aspect of comparative oncology aimed at improving outcomes for dogs and human beings. A summary of this meeting and subsequent discussion are provided to inform the scientific and clinical community of the potential for this initiative. Canine and human comparisons represent an unprecedented opportunity to complement conventional brain tumor research paradigms, addressing a devastating disease for which innovative diagnostic and treatment strategies are clearly needed., (Published by Oxford University Press on behalf of the Society for Neuro-Oncology 2016. This work is written by (a) US Government employee(s) and is in the public domain in the US.)
- Published
- 2016
- Full Text
- View/download PDF
12. A Comparative Oncology Study of Iniparib Defines Its Pharmacokinetic Profile and Biological Activity in a Naturally-Occurring Canine Cancer Model.
- Author
-
Saba C, Paoloni M, Mazcko C, Kisseberth W, Burton JH, Smith A, Wilson-Robles H, Allstadt S, Vail D, Henry C, Lana S, Ehrhart EJ, Charles B, Kent M, Lawrence J, Burgess K, Borgatti A, Suter S, Woods P, Gordon I, Vrignaud P, Khanna C, and LeBlanc AK
- Subjects
- Animals, Antineoplastic Agents therapeutic use, Benzamides therapeutic use, Breast Neoplasms drug therapy, Dogs, Dose-Response Relationship, Drug, Drug Administration Schedule, Female, Male, Maximum Tolerated Dose, Poly(ADP-ribose) Polymerase Inhibitors pharmacokinetics, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Triple Negative Breast Neoplasms drug therapy, Antineoplastic Agents pharmacokinetics, Benzamides pharmacokinetics, Neoplasms drug therapy, Neoplasms veterinary
- Abstract
Development of iniparib as an anti-cancer agent was hindered in part by lingering questions regarding its mechanism of action, the activity of its metabolites, and their potential accumulation in tumors. Due to strong similarities in metabolism of iniparib between humans and dogs, a veterinary clinical trial in pet dogs with spontaneous cancers was designed to answer specific questions pertaining to pharmacokinetic exposures and tolerability of iniparib. Dogs were treated with iniparib alone and in combination with carboplatin chemotherapy. Iniparib doses ranged between 10-70 mg/kg intravenously (IV). Plasma, tumor and normal tissue samples were collected before and at various time points scheduled after exposure for pharmacokinetic and biologic analysis. The primary endpoints included characterization of dose-limiting toxicities (DLT) and determination of the drug exposures that could be achieved in both normal and tumor tissues. Nineteen dogs were treated. DLT included fever, anorexia, diarrhea, neutropenia, and thrombocytopenia; most effects were attributable to carboplatin based on the timing of adverse event onset. The maximum tolerated dose (MTD) of iniparib was not identified. Moderate to high variability in plasma exposure was noted for iniparib and all metabolites between animals. When quantifiable, iniparib and metabolite plasma:tumor ratios were < 0.088 and <1.7, respectively. In this study, iniparib was well tolerated as a single agent and in combination with carboplatin over a range of doses. However, clinically relevant concentrations of the parent drug and selected metabolites were not detectable in canine tumor tissues at any studied dose, thus eliminating expectations for clinical responses in dogs or humans. Negative clinical trials in humans, and the uncertainties of its mechanism of action, ultimately led to the decision to stop clinical development of the drug. Nevertheless, the questions that can be asked and answered within the comparative oncology approach are evident from this successfully executed comparative clinical trial and exemplify the value of such studies in drug development.
- Published
- 2016
- Full Text
- View/download PDF
13. The Establishment of the Pfizer-Canine Comparative Oncology and Genomics Consortium Biospecimen Repository.
- Author
-
Mazcko C and Thomas R
- Abstract
The Canine Comparative Oncology and Genomics Consortium (CCOGC) was formed in 2004 in an effort to capitalize on the generation of a domestic dog genome sequence assembly [1], which created new opportunities to investigate canine cancers at the molecular level [2]. [...].
- Published
- 2015
- Full Text
- View/download PDF
14. Defining the Pharmacodynamic Profile and Therapeutic Index of NHS-IL12 Immunocytokine in Dogs with Malignant Melanoma.
- Author
-
Paoloni M, Mazcko C, Selting K, Lana S, Barber L, Phillips J, Skorupski K, Vail D, Wilson H, Biller B, Avery A, Kiupel M, LeBlanc A, Bernhardt A, Brunkhorst B, Tighe R, and Khanna C
- Subjects
- Animals, Antineoplastic Agents administration & dosage, Antineoplastic Agents pharmacokinetics, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes metabolism, Cytokines blood, Dog Diseases blood, Dogs, Female, Immunoglobulin G administration & dosage, Immunologic Factors administration & dosage, Immunologic Factors pharmacokinetics, Immunophenotyping, Infusions, Subcutaneous, Interleukin-12 administration & dosage, Interleukin-12 pharmacokinetics, Leukocytes, Mononuclear immunology, Leukocytes, Mononuclear metabolism, Lymph Nodes immunology, Lymph Nodes metabolism, Lymph Nodes pathology, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating metabolism, Male, Recombinant Fusion Proteins administration & dosage, Recombinant Fusion Proteins pharmacokinetics, Treatment Outcome, Antineoplastic Agents pharmacology, Dog Diseases drug therapy, Dog Diseases pathology, Immunoglobulin G pharmacology, Immunologic Factors pharmacology, Interleukin-12 pharmacology, Melanoma veterinary, Recombinant Fusion Proteins pharmacology
- Abstract
Background: Interleukin (IL)-12 is a pro-inflammatory cytokine that mediates T-helper type 1 responses and cytotoxic T-cell activation, contributing to its utility as anti-cancer agent. Systemic administration of IL-12 often results in unacceptable toxicity; therefore, strategies to direct delivery of IL-12 to tumors are under investigation. The objective of this study was to assist the preclinical development of NHS-IL12, an immunocytokine consisting of an antibody, which targets necrotic tumor regions, linked to IL-12. Specifically this study sought to evaluate the safety, serum pharmacokinetics, anti-tumor activity, and immune modulation of NHS-IL12 in dogs with naturally occurring cancers., Methodology/principal Findings: A rapid dose-escalation study of NHS-IL12 administered subcutaneously to dogs with melanoma was conducted through the Comparative Oncology Trials Consortium (COTC). Eleven dogs were enrolled in four dose-escalation cohorts; thereafter, an additional seven dogs were treated at the defined tolerable dose of 0.8 mg/m2. The expanded cohort at this fixed dose (ten dogs in total) was accrued for further pharmacokinetics and pharmacodynamics assessment. NHS-IL12 levels, serum cytokine concentrations, and peripheral blood mononuclear cell characterization (post-treatment) and draining lymph node immune profiling, and tumor biopsies (pre- and post-treatment) were collected. Adverse events included thrombocytopenia, liver enzymopathies, fever, and vasculitis. Correlation between interferon (IFN)-γ induction, adverse events, and NHS-IL12 exposure (maximum concentration and area under the concentration-time curve) were dose-dependent. Serum IL-10 levels and intratumoral CD8+ populations increased after treatment. Partial responses, according to Response Evaluation Criteria in Solid Tumors (RECIST) criteria, were observed in two dogs treated with NHS-IL12 0.8 mg/m2 and 1.6 mg/m2., Conclusions/significance: NHS-IL12 was administered safely to dogs with melanoma and both immunologic and clinical activity was observed. This study successfully defined a narrow therapeutic window for systemic delivery of NHS-IL12 via the subcutaneous route. Results will inform the design and implementation of first-in-human clinical trials of NHS-IL12 in cancer patients.
- Published
- 2015
- Full Text
- View/download PDF
15. Prospective molecular profiling of canine cancers provides a clinically relevant comparative model for evaluating personalized medicine (PMed) trials.
- Author
-
Paoloni M, Webb C, Mazcko C, Cherba D, Hendricks W, Lana S, Ehrhart EJ, Charles B, Fehling H, Kumar L, Vail D, Henson M, Childress M, Kitchell B, Kingsley C, Kim S, Neff M, Davis B, Khanna C, and Trent J
- Subjects
- Animals, Cluster Analysis, Computational Biology methods, Disease Models, Animal, Dogs, Female, Genomics methods, Humans, Male, Neoplasms pathology, Prospective Studies, Gene Expression Profiling, Neoplasms genetics, Precision Medicine
- Abstract
Background: Molecularly-guided trials (i.e. PMed) now seek to aid clinical decision-making by matching cancer targets with therapeutic options. Progress has been hampered by the lack of cancer models that account for individual-to-individual heterogeneity within and across cancer types. Naturally occurring cancers in pet animals are heterogeneous and thus provide an opportunity to answer questions about these PMed strategies and optimize translation to human patients. In order to realize this opportunity, it is now necessary to demonstrate the feasibility of conducting molecularly-guided analysis of tumors from dogs with naturally occurring cancer in a clinically relevant setting., Methodology: A proof-of-concept study was conducted by the Comparative Oncology Trials Consortium (COTC) to determine if tumor collection, prospective molecular profiling, and PMed report generation within 1 week was feasible in dogs. Thirty-one dogs with cancers of varying histologies were enrolled. Twenty-four of 31 samples (77%) successfully met all predefined QA/QC criteria and were analyzed via Affymetrix gene expression profiling. A subsequent bioinformatics workflow transformed genomic data into a personalized drug report. Average turnaround from biopsy to report generation was 116 hours (4.8 days). Unsupervised clustering of canine tumor expression data clustered by cancer type, but supervised clustering of tumors based on the personalized drug report clustered by drug class rather than cancer type., Conclusions: Collection and turnaround of high quality canine tumor samples, centralized pathology, analyte generation, array hybridization, and bioinformatic analyses matching gene expression to therapeutic options is achievable in a practical clinical window (<1 week). Clustering data show robust signatures by cancer type but also showed patient-to-patient heterogeneity in drug predictions. This lends further support to the inclusion of a heterogeneous population of dogs with cancer into the preclinical modeling of personalized medicine. Future comparative oncology studies optimizing the delivery of PMed strategies may aid cancer drug development.
- Published
- 2014
- Full Text
- View/download PDF
16. Rapamycin pharmacokinetic and pharmacodynamic relationships in osteosarcoma: a comparative oncology study in dogs.
- Author
-
Paoloni MC, Mazcko C, Fox E, Fan T, Lana S, Kisseberth W, Vail DM, Nuckolls K, Osborne T, Yalkowsy S, Gustafson D, Yu Y, Cao L, and Khanna C
- Subjects
- Animals, Cell Line, Tumor, Cohort Studies, Dog Diseases metabolism, Dogs, Dose-Response Relationship, Drug, Female, Immunosuppressive Agents administration & dosage, Injections, Intramuscular, Male, Osteosarcoma metabolism, Osteosarcoma pathology, Sirolimus administration & dosage, Dog Diseases pathology, Immunosuppressive Agents pharmacokinetics, Immunosuppressive Agents pharmacology, Osteosarcoma veterinary, Sirolimus pharmacokinetics, Sirolimus pharmacology
- Abstract
Background: Signaling through the mTOR pathway contributes to growth, progression and chemoresistance of several cancers. Accordingly, inhibitors have been developed as potentially valuable therapeutics. Their optimal development requires consideration of dose, regimen, biomarkers and a rationale for their use in combination with other agents. Using the infrastructure of the Comparative Oncology Trials Consortium many of these complex questions were asked within a relevant population of dogs with osteosarcoma to inform the development of mTOR inhibitors for future use in pediatric osteosarcoma patients., Methodology/principal Findings: This prospective dose escalation study of a parenteral formulation of rapamycin sought to define a safe, pharmacokinetically relevant, and pharmacodynamically active dose of rapamycin in dogs with appendicular osteosarcoma. Dogs entered into dose cohorts consisting of 3 dogs/cohort. Dogs underwent a pre-treatment tumor biopsy and collection of baseline PBMC. Dogs received a single intramuscular dose of rapamycin and underwent 48-hour whole blood pharmacokinetic sampling. Additionally, daily intramuscular doses of rapamycin were administered for 7 days with blood rapamycin trough levels collected on Day 8, 9 and 15. At Day 8 post-treatment collection of tumor and PBMC were obtained. No maximally tolerated dose of rapamycin was attained through escalation to the maximal planned dose of 0.08 mg/kg (2.5 mg/30 kg dog). Pharmacokinetic analysis revealed a dose-dependent exposure. In all cohorts modulation of the mTOR pathway in tumor and PBMC (pS6RP/S6RP) was demonstrated. No change in pAKT/AKT was seen in tumor samples following rapamycin therapy., Conclusions/significance: Rapamycin may be safely administered to dogs and can yield therapeutic exposures. Modulation pS6RP/S6RP in tumor tissue and PBMCs was not dependent on dose. Results from this study confirm that the dog may be included in the translational development of rapamycin and potentially other mTOR inhibitors. Ongoing studies of rapamycin in dogs will define optimal schedules for their use in cancer and evaluate the role of rapamycin use in the setting of minimal residual disease.
- Published
- 2010
- Full Text
- View/download PDF
17. Guiding the optimal translation of new cancer treatments from canine to human cancer patients.
- Author
-
Khanna C, London C, Vail D, Mazcko C, and Hirschfeld S
- Subjects
- Animals, Clinical Trials as Topic statistics & numerical data, Clinical Trials as Topic veterinary, Dogs, Drug Discovery, Humans, Risk Factors, Antineoplastic Agents therapeutic use, Disease Models, Animal, Neoplasms drug therapy, Neoplasms veterinary, Practice Guidelines as Topic
- Abstract
On June 20, 2008, a meeting entitled "Translation of new cancer treatments from canine to human cancer patients," sponsored by the National Cancer Institute in Bethesda, Maryland, was convened to discuss the potential value, opportunity, risks, and rewards of an integrated and comparative drug development path for new cancer therapeutics that includes naturally occurring cancers in pet animals. A summary of this meeting and subsequent discussion are provided here to afford clarity on the conduct of these studies so as to optimize the opportunities provided by this novel drug development and modeling strategy.
- Published
- 2009
- Full Text
- View/download PDF
18. Launching a novel preclinical infrastructure: comparative oncology trials consortium directed therapeutic targeting of TNFalpha to cancer vasculature.
- Author
-
Paoloni MC, Tandle A, Mazcko C, Hanna E, Kachala S, Leblanc A, Newman S, Vail D, Henry C, Thamm D, Sorenmo K, Hajitou A, Pasqualini R, Arap W, Khanna C, and Libutti SK
- Subjects
- Adenoviridae genetics, Animals, Dogs, Genetic Therapy methods, Genetic Vectors, Neoplasms blood supply, Neoplasms veterinary, Oligopeptides administration & dosage, Treatment Outcome, Tumor Necrosis Factor-alpha therapeutic use, Neoplasms therapy, Neovascularization, Pathologic drug therapy, Tumor Necrosis Factor-alpha administration & dosage
- Abstract
Background: Under the direction and sponsorship of the National Cancer Institute, we report on the first pre-clinical trial of the Comparative Oncology Trials Consortium (COTC). The COTC is a novel infrastructure to integrate cancers that naturally develop in pet dogs into the development path of new human drugs. Trials are designed to address questions challenging in conventional preclinical models and early phase human trials. Large animal spontaneous cancer models can be a valuable addition to successful studies of cancer biology and novel therapeutic drug, imaging and device development., Methodology/principal Findings: Through this established infrastructure, the first trial of the COTC (COTC001) evaluated a targeted AAV-phage vector delivering tumor necrosis factor (RGD-A-TNF) to alphaV integrins on tumor endothelium. Trial progress and data was reviewed contemporaneously using a web-enabled electronic reporting system developed for the consortium. Dose-escalation in cohorts of 3 dogs (n = 24) determined an optimal safe dose (5x10(12) transducing units intravenous) of RGD-A-TNF. This demonstrated selective targeting of tumor-associated vasculature and sparing of normal tissues assessed via serial biopsy of both tumor and normal tissue. Repetitive dosing in a cohort of 14 dogs, at the defined optimal dose, was well tolerated and led to objective tumor regression in two dogs (14%), stable disease in six (43%), and disease progression in six (43%) via Response Evaluation Criteria in Solid Tumors (RECIST)., Conclusions/significance: The first study of the COTC has demonstrated the utility and efficiency of the established infrastructure to inform the development of new cancer drugs within large animal naturally occurring cancer models. The preclinical evaluation of RGD-A-TNF within this network provided valuable and necessary data to complete the design of first-in-man studies.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.