1
|
Seung BJ, Sur JH. Detection of PIK3CA hotspot mutations in canine mammary tumors using droplet digital PCR: tissue validation and liquid biopsy feasibility. Sci Rep 2024; 14:25587. [PMID: 39462049 PMCID: PMC11512996 DOI: 10.1038/s41598-024-76820-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Domestic dogs (Canis lupus familiaris) serve as valuable translational models for human cancer research due to their biological similarities. Canine mammary tumors (CMTs), frequently diagnosed in female dogs, share various characteristics with human breast cancers. This study investigates the PIK3CA (H1047R) mutation in CMTs using droplet digital PCR (ddPCR) and explores the potential of liquid biopsy for non-invasive detection. We analyzed 80 formalin-fixed, paraffin-embedded (FFPE) CMT tissue samples and compared ddPCR results with next-generation sequencing (NGS) data, achieving high concordance. Plasma and serum samples were also assessed for mutation concordance with tissue results. Our findings indicate a higher frequency of the PIK3CA (H1047R) mutations in benign and grade I malignant CMTs compared to more aggressive malignancies. The ddPCR assay demonstrated high sensitivity and specificity, with plasma testing showing 78.6% sensitivity and 87.5% specificity, and serum testing showing 66.7% sensitivity and 90.0% specificity. These results highlight the viability of liquid biopsy as a minimally invasive method for monitoring PIK3CA mutations in canine patients. The study suggests that liquid biopsy techniques hold significant promise for improving the early detection and monitoring of canine cancers, warranting further research to refine these methods and explore their applications in canine cancer diagnostics and treatment.
Collapse
Affiliation(s)
- Byung-Joon Seung
- Department of Veterinary Pathology, College of Veterinary Medicine, Konkuk University, Seoul, 05029, South Korea.
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61802, USA.
| | - Jung-Hyang Sur
- Department of Veterinary Pathology, College of Veterinary Medicine, Konkuk University, Seoul, 05029, South Korea.
- Komipharm International Co., Ltd., Siheung-si, Gyonggi-do, 15094, South Korea.
| |
Collapse
|
2
|
Seung BJ, Bae MK, Sur JH. Regional Variations in and Key Predictors of Feline Tumor Malignancy: A Decade-Long Retrospective Study in Korea. Animals (Basel) 2024; 14:2989. [PMID: 39457919 PMCID: PMC11503808 DOI: 10.3390/ani14202989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/11/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Feline cancer is increasingly recognized as a major cause of mortality, yet data on tumor prevalence and behavior in cats, particularly in non-Western regions, remain limited. This study analyzed a decade of feline tumor data in Korea from 2012 to 2022, focusing on age, breed, and anatomical location as predictors of malignancy. Data were collected from 683 cats, with regression analysis applied to determine significant associations. Older cats exhibited a markedly higher risk of malignancy, particularly in mast cell and mammary tumors. Tumors in the mammary gland and alimentary tract had malignancy rates exceeding 90%, underscoring the need for early detection in these regions. Interestingly, squamous cell carcinoma was rare in the skin, in stark contrast to Western studies, likely reflecting differences in environmental exposure. While breed was not a statistically significant predictor, certain breeds, including Persians and Russian Blues, showed a higher frequency of malignancy. These findings highlight the importance of regional tumor research in cats and the need for larger, multicenter datasets that incorporate environmental, genetic, and lifestyle factors. Understanding these influences will help refine veterinary care and improve cancer treatment outcomes in feline populations.
Collapse
Affiliation(s)
- Byung-Joon Seung
- Department of Veterinary Pathology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea;
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61802, USA
| | - Min-Kyung Bae
- Department of Veterinary Pathology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea;
- Research Institute, Green Vet, Yongin-si 16907, Republic of Korea
| | - Jung-Hyang Sur
- Department of Veterinary Pathology, College of Veterinary Medicine, Konkuk University, Seoul 05029, Republic of Korea;
- Komipharm International Co., Ltd., Siheung-si 15094, Republic of Korea
| |
Collapse
|
3
|
Guo S, Zheng S, Liu M, Wang G. Novel Anti-Cancer Stem Cell Compounds: A Comprehensive Review. Pharmaceutics 2024; 16:1024. [PMID: 39204369 PMCID: PMC11360402 DOI: 10.3390/pharmaceutics16081024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Cancer stem cells (CSCs) possess a significant ability to renew themselves, which gives them a strong capacity to form tumors and expand to encompass additional body areas. In addition, they possess inherent resistance to chemotherapy and radiation therapies used to treat many forms of cancer. Scientists have focused on investigating the signaling pathways that are highly linked to the ability of CSCs to renew themselves and maintain their stem cell properties. The pathways encompassed are Notch, Wnt/β-catenin, hedgehog, STAT3, NF-κB, PI-3K/Akt/mTOR, sirtuin, ALDH, MDM2, and ROS. Recent studies indicate that directing efforts towards CSC cells is essential in eradicating the overall cancer cell population and reducing the likelihood of tumor metastasis. As our comprehension of the mechanisms that stimulate CSC activity, growth, and resistance to chemotherapy advances, the discovery of therapeutic drugs specifically targeting CSCs, such as small-molecule compounds, holds the potential to revolutionize cancer therapy. This review article examines and analyzes the novel anti-CSC compounds that have demonstrated effective and selective targeting of pathways associated with the renewal and stemness of CSCs. We also discussed their special drug metabolism and absorption mechanisms. CSCs have been the subject of much study in cancer biology. As a possible treatment for malignancies, small-molecule drugs that target CSCs are gaining more and more attention. This article provides a comprehensive review of the current state of key small-molecule compounds, summarizes their recent developments, and anticipates the future discovery of even more potent and targeted compounds, opening up new avenues for cancer treatment.
Collapse
Affiliation(s)
- Shanchun Guo
- RCMI Cancer Research Center and Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA;
| | - Shilong Zheng
- RCMI Cancer Research Center and Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA;
| | - Mingli Liu
- Department of Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA;
| | - Guangdi Wang
- RCMI Cancer Research Center and Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA;
| |
Collapse
|
4
|
Wu K, Rodrigues L, Post G, Harvey G, White M, Miller A, Lambert L, Lewis B, Lopes C, Zou J. Analyses of canine cancer mutations and treatment outcomes using real-world clinico-genomics data of 2119 dogs. NPJ Precis Oncol 2023; 7:8. [PMID: 36658200 PMCID: PMC9852553 DOI: 10.1038/s41698-023-00346-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 01/05/2023] [Indexed: 01/21/2023] Open
Abstract
Spontaneous tumors in canines share significant genetic and histological similarities with human tumors, positioning them as valuable models to guide drug development. However, current translational studies have limited real world evidence as cancer outcomes are dispersed across veterinary clinics and genomic tests are rarely performed on dogs. In this study, we aim to expand the value of canine models by systematically characterizing genetic mutations in tumors and their response to targeted treatments. In total, we collect and analyze survival outcomes for 2119 tumor-bearing dogs and the prognostic effect of genomic alterations in a subset of 1108 dogs. Our analysis identifies prognostic concordance between canines and humans in several key oncogenes, including TP53 and PIK3CA. We also find that several targeted treatments designed for humans are associated with a positive prognosis when used to treat canine tumors with specific genomic alterations, underscoring the value of canine models in advancing drug discovery for personalized oncology.
Collapse
Affiliation(s)
- Kevin Wu
- One Health Company, Palo Alto, CA, US
- Department of Biomedical Data Science, Stanford University, Stanford, US
| | | | | | | | | | | | | | | | | | - James Zou
- One Health Company, Palo Alto, CA, US
- Department of Biomedical Data Science, Stanford University, Stanford, US
| |
Collapse
|
5
|
Delgado M, Buffington CT, Bain M, Smith DL, Vernau K. Early maternal separation is not associated with changes in telomere length in domestic kittens ( Felis catus). PeerJ 2021; 9:e11394. [PMID: 34141465 PMCID: PMC8176934 DOI: 10.7717/peerj.11394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Accepted: 04/12/2021] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE Studies of multiple species have found that adverse early life experiences, including childhood trauma and maternal separation, can result in accelerated telomere shortening. The objective of this study was to determine if premature separation from the mother affected telomere length in domestic kittens (Felis catus). Subjects were 42 orphaned kittens and 10 mother-reared kittens from local animal rescue groups and shelters. DNA was extracted from whole blood collected from kittens at approximately 1 week and 2 months of age. Telomere length was assessed by qPCR (quantitative polymerase chain reaction) from a total of 86 samples and expressed as a ratio of telomere PCR relative to a single copy gene PCR (T/S). RESULTS A generalized linear mixed model found there were no detectable differences in telomere length based on survival (F 1, 76.2 = 3.35, p = 0.07), orphan status (F 1, 56.5 = 0.44, p = 0.51), time point (F 1, 43.5 = 0.19, p = 0.67), or the interaction between orphan status and time (F 1, 43.5 = 0.86, p = 0.36). Although in other species telomere shortening is commonly associated with aging, even early in life, we did not find evidence for telomere shortening by two months of age. Our results suggest that the experience of early maternal separation in domestic cats who are subsequently hand-reared by humans does not accelerate telomere shortening compared to mother-reared kittens, at least in the first few months of life.
Collapse
Affiliation(s)
- Mikel Delgado
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States of America
| | - C.A. Tony Buffington
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States of America
| | - Melissa Bain
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States of America
| | - Dana L. Smith
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA, United States of America
| | - Karen Vernau
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, Davis, CA, United States of America
| |
Collapse
|
6
|
Chibuk J, Flory A, Kruglyak KM, Leibman N, Nahama A, Dharajiya N, van den Boom D, Jensen TJ, Friedman JS, Shen MR, Clemente-Vicario F, Chorny I, Tynan JA, Lytle KM, Holtvoigt LE, Murtaza M, Diaz LA, Tsui DWY, Grosu DS. Horizons in Veterinary Precision Oncology: Fundamentals of Cancer Genomics and Applications of Liquid Biopsy for the Detection, Characterization, and Management of Cancer in Dogs. Front Vet Sci 2021; 8:664718. [PMID: 33834049 PMCID: PMC8021921 DOI: 10.3389/fvets.2021.664718] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 02/23/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer is the leading cause of death in dogs, in part because many cases are identified at an advanced stage when clinical signs have developed, and prognosis is poor. Increased understanding of cancer as a disease of the genome has led to the introduction of liquid biopsy testing, allowing for detection of genomic alterations in cell-free DNA fragments in blood to facilitate earlier detection, characterization, and management of cancer through non-invasive means. Recent discoveries in the areas of genomics and oncology have provided a deeper understanding of the molecular origins and evolution of cancer, and of the "one health" similarities between humans and dogs that underlie the field of comparative oncology. These discoveries, combined with technological advances in DNA profiling, are shifting the paradigm for cancer diagnosis toward earlier detection with the goal of improving outcomes. Liquid biopsy testing has already revolutionized the way cancer is managed in human medicine - and it is poised to make a similar impact in veterinary medicine. Multiple clinical use cases for liquid biopsy are emerging, including screening, aid in diagnosis, targeted treatment selection, treatment response monitoring, minimal residual disease detection, and recurrence monitoring. This review article highlights key scientific advances in genomics and their relevance for veterinary oncology, with the goal of providing a foundational introduction to this important topic for veterinarians. As these technologies migrate from human medicine into veterinary medicine, improved awareness and understanding will facilitate their rapid adoption, for the benefit of veterinary patients.
Collapse
Affiliation(s)
| | | | | | - Nicole Leibman
- The Cancer Institute, Animal Medical Center, New York, NY, United States
| | | | | | | | | | | | - M. Richard Shen
- RS Technology Ventures LLC., Rancho Santa Fe, CA, United States
| | | | | | | | | | | | - Muhammed Murtaza
- Department of Surgery and Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI, United States
| | - Luis A. Diaz
- Division of Solid Tumor Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | | | | |
Collapse
|
7
|
Altamura G, Degli Uberti B, Galiero G, De Luca G, Power K, Licenziato L, Maiolino P, Borzacchiello G. The Small Molecule BIBR1532 Exerts Potential Anti-cancer Activities in Preclinical Models of Feline Oral Squamous Cell Carcinoma Through Inhibition of Telomerase Activity and Down-Regulation of TERT. Front Vet Sci 2021; 7:620776. [PMID: 33553285 PMCID: PMC7855307 DOI: 10.3389/fvets.2020.620776] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/07/2020] [Indexed: 11/26/2022] Open
Abstract
Expression of telomerase reverse transcriptase (TERT) and telomerase activity (TA) is a main feature of cancer, contributing to cell immortalization by causing telomeres dysfunction. BIBR1532 is a potent telomerase inhibitor that showed potential anti-tumor activities in several types of cancer, by triggering replicative senescence and apoptosis. In a previous work, we detected, for the first time, TERT expression and TA in preclinical models of feline oral squamous cell carcinoma (FOSCC); therefore, we aimed at extending our investigation by testing the effects of treatment with BIBR1532, in order to explore the role of telomerase in this tumor and foreshadow the possibility of it being considered as a future therapeutic target. In the present study, treatment of FOSCC cell lines SCCF1, SCCF2, and SCCF3 with BIBR1532 resulted in successful inhibition of TA, with subsequent cell growth stoppage and decrease in cell viability. Molecular data showed that up-regulation of cell cycle inhibitor p21, unbalancing of Bax/Bcl-2 ratio, and down-regulation of survival gene Survivin were mostly involved in the observed cellular events. Moreover, BIBR1532 diminished the expression of TERT and its transcriptional activator cMyc, resulting in the down-regulation of epidermal growth factor receptor (EGFR), phospho-ERK/ERK ratio, and matrix metalloproteinases (MMPs)-1/-2 and−9, likely as a consequence of an impairment of TERT extra-telomeric functions. Taken together, our data suggest that BIBR1532 exerts multiple anti-cancer activities in FOSCC by inhibiting telomerase pathway and interfering with signaling routes involved in cell proliferation, cell survival, and invasion, paving the way for future translational studies aimed at evaluating its possible employment in the treatment of this severe tumor of cats.
Collapse
Affiliation(s)
- Gennaro Altamura
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | | | - Giorgio Galiero
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, Portici, Naples, Italy
| | - Giovanna De Luca
- Istituto Zooprofilattico Sperimentale del Mezzogiorno, Portici, Naples, Italy
| | - Karen Power
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Luca Licenziato
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Paola Maiolino
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Giuseppe Borzacchiello
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| |
Collapse
|
8
|
Robertson N, Schook LB, Schachtschneider KM. Porcine cancer models: potential tools to enhance cancer drug trials. Expert Opin Drug Discov 2020; 15:893-902. [PMID: 32378979 DOI: 10.1080/17460441.2020.1757644] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The amount of time and money invested into cancer drug research, development, and clinical trials has continually increased over the past few decades. Despite record high cancer drug approval rates, cancer remains a leading cause of death. This suggests the need for more effective tools to help bring novel therapies to clinical practice in a timely manner. AREAS COVERED In this review, current issues associated with clinical trials are discussed, specifically focusing on poor accrual rates and time for trial completion. In addition, details regarding preclinical studies required before advancing to clinical trials are discussed, including advantages and limitations of current preclinical animal cancer models and their relevance to human cancer trials. Finally, new translational porcine cancer models (Oncopig Cancer Model (OCM)) are presented as potential co-clinical trial models. EXPERT OPINION In order to address issues impacting the poor success rate of oncology clinical trials, we propose the incorporation of the transformative OCM 'co-clinical trial' pathway into the cancer drug approval process. Due to the Oncopig's high homology to humans and similar tumor phenotypes, their utilization can provide improved preclinical prediction of both drug safety and efficacy prior to investing significant time and money in human clinical trials.
Collapse
Affiliation(s)
- Noah Robertson
- Department of Radiology, University of Illinois at Chicago , Chicago, IL, USA
| | - Lawrence B Schook
- Department of Radiology, University of Illinois at Chicago , Chicago, IL, USA.,Department of Animal Sciences, University of Illinois at Urbana-Champaign , Urbana, IL, USA
| | - Kyle M Schachtschneider
- Department of Radiology, University of Illinois at Chicago , Chicago, IL, USA.,Department of Biochemistry & Molecular Genetics, University of Illinois at Chicago , Chicago, IL, USA
| |
Collapse
|
9
|
Gray M, Meehan J, Martínez-Pérez C, Kay C, Turnbull AK, Morrison LR, Pang LY, Argyle D. Naturally-Occurring Canine Mammary Tumors as a Translational Model for Human Breast Cancer. Front Oncol 2020; 10:617. [PMID: 32411603 PMCID: PMC7198768 DOI: 10.3389/fonc.2020.00617] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 04/03/2020] [Indexed: 01/03/2023] Open
Abstract
Despite extensive research over many decades, human breast cancer remains a major worldwide health concern. Advances in pre-clinical and clinical research has led to significant improvements in recent years in how we manage breast cancer patients. Although survival rates of patients suffering from localized disease has improved significantly, the prognosis for patients diagnosed with metastatic disease remains poor with 5-year survival rates at only 25%. In vitro studies using immortalized cell lines and in vivo mouse models, typically using xenografted cell lines or patient derived material, are commonly used to study breast cancer. Although these techniques have undoubtedly increased our molecular understanding of breast cancer, these research models have significant limitations and have contributed to the high attrition rates seen in cancer drug discovery. It is estimated that only 3-6% of drugs that show promise in these pre-clinical models will reach clinical use. Models that can reproduce human breast cancer more accurately are needed if significant advances are to be achieved in improving cancer drug research, treatment outcomes, and prognosis. Canine mammary tumors are a naturally-occurring heterogenous group of cancers that have several features in common with human breast cancer. These similarities include etiology, signaling pathway activation and histological classification. In this review article we discuss the use of naturally-occurring canine mammary tumors as a translational animal model for human breast cancer research.
Collapse
Affiliation(s)
- Mark Gray
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - James Meehan
- Translational Oncology Research Group, Cancer Research UK Edinburgh Center, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Carlos Martínez-Pérez
- Translational Oncology Research Group, Cancer Research UK Edinburgh Center, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Charlene Kay
- Translational Oncology Research Group, Cancer Research UK Edinburgh Center, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Arran K Turnbull
- Translational Oncology Research Group, Cancer Research UK Edinburgh Center, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Linda R Morrison
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Lisa Y Pang
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - David Argyle
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
10
|
Altamura G, Martano M, Licenziato L, Maiolino P, Borzacchiello G. Telomerase Reverse Transcriptase (TERT) Expression, Telomerase Activity, and Expression of Matrix Metalloproteinases (MMP)-1/-2/-9 in Feline Oral Squamous Cell Carcinoma Cell Lines Associated With Felis catus Papillomavirus Type-2 Infection. Front Vet Sci 2020; 7:148. [PMID: 32292795 PMCID: PMC7118734 DOI: 10.3389/fvets.2020.00148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 02/28/2020] [Indexed: 12/11/2022] Open
Abstract
Telomerase activity contributes to cell immortalization by avoiding telomere shortening at each cell division; indeed, its catalytic subunit telomerase reverse transcriptase (TERT) is overexpressed in many tumors, including human oral squamous cell carcinoma (hOSCC). In these tumors, matrix metalloproteinases (MMPs), a group of zinc-dependent endopeptidases involved in cell migration, contribute to invasive potential of cancer cells. A proportion of hOSCC is associated with infection by high-risk human papillomavirus (HR-HPVs), whose E6 oncogene enhances TERT and MMPs expression, thus promoting cancer progression. Feline oral squamous cell carcinoma (FOSCC) is a malignant tumor with highly invasive phenotype; however, studies on telomerase activity, TERT, and MMPs expression are scarce. In this study, we demonstrate telomerase activity, expression of TERT, and its transcriptional activator cMyc along with expression of MMP-1, -2, and -9 in FOSCC-derived cell lines SCCF2 and SCCF3, suggesting a contribution by these pathways in cell immortalization and invasion in these tumors. Recent studies suggest that a sub-group of FOSCC as well as SCCF2 and SCCF3 are associated with Felis catus PV type-2 (FcaPV-2) infection. However, in this work, FcaPV-2 E6 gene knock-down caused no shift in either TERT, cMyc, or MMPs levels, suggesting that, unlike its human counterpart, the viral oncogene plays no role in their regulation.
Collapse
Affiliation(s)
- Gennaro Altamura
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Manuela Martano
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Luca Licenziato
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Paola Maiolino
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| | - Giuseppe Borzacchiello
- Department of Veterinary Medicine and Animal Productions, University of Naples Federico II, Naples, Italy
| |
Collapse
|
11
|
Lorke M, Willen M, Lucas K, Schille JT, Lüder Ripoli F, Willenbrock S, Beyerbach M, Wefstaedt P, Murua Escobar H, Nolte I. Effect of antioxidants, mitochondrial cofactors and omega-3 fatty acids on telomere length and kinematic joint mobility in young and old shepherd dogs - A randomized, blinded and placebo-controlled study. Res Vet Sci 2020; 129:137-153. [PMID: 32000015 DOI: 10.1016/j.rvsc.2020.01.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 11/23/2019] [Accepted: 01/08/2020] [Indexed: 12/18/2022]
Abstract
In dogs, decreasing telomere length is a biomarker for cellular aging. On a systemic level, aging affects the locomotor system in particular, leading to restricted joint mobility. As aging is thought to be related to oxidative stress, it may be counteracted by a diet enriched with antioxidants, mitochondrial cofactors and omega-3 fatty acids. This randomized, blinded and placebo-controlled study examined the influence of an accordingly enriched diet compared to a control diet on 36 young and 38 old shepherd dogs. At the outset, after 3 and after 6 months, mean and minimum telomere lengths were measured. Furthermore, minimum and maximum joint angles and range of motion of the shoulder, elbow, carpal, hip, stifle and tarsal joints were measured by computer-assisted gait analysis. A positive influence of the enriched diet on old dogs could be verified for minimum telomere length and all three parameters of the shoulder joint on the side with the higher vertical ground reaction force after 6 months. In the other joints there were less significant differences; in some cases they indicated a contrary influence of the enriched diet on young dogs, probably due to its reduced protein content. The greater effect of the enriched diet on minimum than on mean telomere length may be due to the higher preference of telomerase for short telomeres. The greater effect on shoulder joint mobility is explained by the greater influence of musculature and connective tissue in this joint. For elderly dogs it is advisable to feed these nutritional supplements.
Collapse
Affiliation(s)
- Malin Lorke
- Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, D-30559 Hannover, Germany
| | - Maray Willen
- Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, D-30559 Hannover, Germany
| | - Karin Lucas
- Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, D-30559 Hannover, Germany
| | - Jan Torben Schille
- Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, D-30559 Hannover, Germany
| | - Florenza Lüder Ripoli
- Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, D-30559 Hannover, Germany; Division of Medicine Clinic III, Hematology, Oncology and Palliative Medicine, University of Rostock, D-18057 Rostock, Germany
| | - Saskia Willenbrock
- Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, D-30559 Hannover, Germany
| | - Martin Beyerbach
- Institute for Biometry, Epidemiology and Information Processing, University of Veterinary Medicine Hannover, Foundation, D-30559 Hannover, Germany
| | - Patrick Wefstaedt
- Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, D-30559 Hannover, Germany
| | - Hugo Murua Escobar
- Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, D-30559 Hannover, Germany; Division of Medicine Clinic III, Hematology, Oncology and Palliative Medicine, University of Rostock, D-18057 Rostock, Germany
| | - Ingo Nolte
- Small Animal Clinic, University of Veterinary Medicine Hannover, Foundation, D-30559 Hannover, Germany.
| |
Collapse
|
12
|
Gray ME, Sullivan P, Marland JRK, Greenhalgh SN, Meehan J, Gregson R, Clutton RE, Cousens C, Griffiths DJ, Murray A, Argyle D. A Novel Translational Ovine Pulmonary Adenocarcinoma Model for Human Lung Cancer. Front Oncol 2019; 9:534. [PMID: 31316911 PMCID: PMC6611418 DOI: 10.3389/fonc.2019.00534] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 06/03/2019] [Indexed: 11/13/2022] Open
Abstract
In vitro cell line and in vivo murine models have historically dominated pre-clinical cancer research. These models can be expensive and time consuming and lead to only a small percentage of anti-cancer drugs gaining a license for human use. Large animal models that reflect human disease have high translational value; these can be used to overcome current pre-clinical research limitations through the integration of drug development techniques with surgical procedures and anesthetic protocols, along with emerging fields such as implantable medical devices. Ovine pulmonary adenocarcinoma (OPA) is a naturally-occurring lung cancer that is caused by the jaagsiekte sheep retrovirus. The disease has similar histological classification and oncogenic pathway activation to that of human lung adenocarcinomas making it a valuable model for studying human lung cancer. Developing OPA models to include techniques used in the treatment of human lung cancer would enhance its translational potential, making it an excellent research tool in assessing cancer therapeutics. In this study we developed a novel OPA model to validate the ability of miniaturized implantable O2 and pH sensors to monitor the tumor microenvironment. Naturally-occurring pre-clinical OPA cases were obtained through an on-farm ultrasound screening programme. Sensors were implanted into OPA tumors of anesthetized sheep using a CT-guided trans-thoracic percutaneous implantation procedure. This study reports the findings from 9 sheep that received sensor implantations. Time taken from initial CT scans to the placement of a single sensor into an OPA tumor was 45 ± 5 min, with all implantations resulting in the successful delivery of sensors into tumors. Immediate post-implantation mild pneumothoraces occurred in 4 sheep, which was successfully managed in all cases. This is, to the best of our knowledge, the first description of the use of naturally-occurring OPA cases as a pre-clinical surgical model. Through the integration of techniques used in the treatment of human lung cancer patients, including ultrasound, general anesthesia, CT and surgery into the OPA model, we have demonstrated its translational potential. Although our research was tailored specifically for the implantation of sensors into lung tumors, we believe the model could also be developed for other pre-clinical applications.
Collapse
Affiliation(s)
- Mark E Gray
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, United Kingdom.,Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Paul Sullivan
- School of Engineering, Institute for Integrated Micro and Nano Systems, Edinburgh, United Kingdom
| | - Jamie R K Marland
- School of Engineering, Institute for Integrated Micro and Nano Systems, Edinburgh, United Kingdom
| | - Stephen N Greenhalgh
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, United Kingdom
| | - James Meehan
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom.,Institute of Sensors, Signals and Systems, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom
| | - Rachael Gregson
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, United Kingdom
| | - R Eddie Clutton
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, United Kingdom
| | - Chris Cousens
- Moredun Research Institute, Pentlands Science Park, Midlothian, United Kingdom
| | - David J Griffiths
- Moredun Research Institute, Pentlands Science Park, Midlothian, United Kingdom
| | - Alan Murray
- School of Engineering, Institute for Integrated Micro and Nano Systems, Edinburgh, United Kingdom
| | - David Argyle
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, Edinburgh, United Kingdom
| |
Collapse
|
13
|
Zhou W, Guo S, Liu M, Burow ME, Wang G. Targeting CXCL12/CXCR4 Axis in Tumor Immunotherapy. Curr Med Chem 2019; 26:3026-3041. [PMID: 28875842 PMCID: PMC5949083 DOI: 10.2174/0929867324666170830111531] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 05/08/2017] [Accepted: 06/14/2017] [Indexed: 12/14/2022]
Abstract
Chemokines, which have chemotactic abilities, are comprised of a family of small cytokines with 8-10 kilodaltons. Chemokines work in immune cells by trafficking and regulating cell proliferation, migration, activation, differentiation, and homing. CXCR-4 is an alpha-chemokine receptor specific for stromal-derived-factor-1 (SDF-1, also known as CXCL12), which has been found to be expressed in more than 23 different types of cancers. Recently, the SDF-1/CXCR-4 signaling pathway has emerged as a potential therapeutic target for human tumor because of its critical role in tumor initiation and progression by activating multiple signaling pathways, such as ERK1/2, ras, p38 MAPK, PLC/ MAPK, and SAPK/ JNK, as well as regulating cancer stem cells. CXCL12/CXCR4 antagonists have been produced, which have shown encouraging results in anti-cancer activity. Here, we provide a brief overview of the CXCL12/CXCR4 axis as a molecular target for cancer treatment. We also review the potential utility of targeting CXCL12/CXCR4 axis in combination of immunotherapy and/or chemotherapy based on up-to-date literature and ongoing research progress.
Collapse
Affiliation(s)
- Weiqiang Zhou
- Key Laboratory of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No.146 North Huanghe St, Huanggu District, Shenyang, Liaoning Province 110034, P. R. China
| | - Shanchun Guo
- RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA
| | - Mingli Liu
- Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | - Matthew E. Burow
- Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Guangdi Wang
- RCMI Cancer Research Center, Xavier University of Louisiana, New Orleans, LA 70125, USA
- Department of Chemistry, Xavier University of Louisiana, New Orleans, LA 70125, USA
| |
Collapse
|
14
|
Ryu JE, Park HK, Choi HJ, Lee HB, Lee HJ, Lee H, Yu ES, Son WC. Expression of the glutamine metabolism-related proteins glutaminase 1 and glutamate dehydrogenase in canine mammary tumours. Vet Comp Oncol 2017; 16:239-245. [PMID: 29266697 DOI: 10.1111/vco.12369] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 10/02/2017] [Accepted: 10/13/2017] [Indexed: 01/14/2023]
Abstract
Glutamine metabolism is an important metabolic pathway for cancer cell survival, and there is a critical connection between tumour growth and glutamine metabolism. Because of their similarities, canine mammary carcinomas are useful for studying human breast cancer. Accordingly, we investigated the correlations between the expression of glutamine metabolism-related proteins and the pathological features of canine mammary tumours. We performed immunohistochemical and western blot analysis of 39 mammary tumour tissues. In immunohistochemical analysis, the expression of glutaminase 1 (GLS1) in the epithelial region increased according to the histological grade (P < .005). In the stromal region, complex-type tumours displayed significantly higher GLS1 intensity than simple-type tumours. However, glutamate dehydrogenase expression did not show the same tendencies as GLS1. The western blot results were consistent with the immunohistochemical findings. These results suggest that the expression of GLS1 is correlates with clinicopathological factors in canine mammary tumours and shows a similar pattern to human breast cancer.
Collapse
Affiliation(s)
- J-E Ryu
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - H-K Park
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - H-J Choi
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - H-B Lee
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - H-J Lee
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea
| | - H Lee
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea.,Pharma R&D Division, GeneOne Life Science, Inc., Seoul, Republic of Korea
| | - E-S Yu
- Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - W-C Son
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, Republic of Korea.,Department of Pathology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| |
Collapse
|
15
|
Marques O, Canadas A, Faria F, Oliveira E, Amorim I, Seixas F, Gama A, Lobo-da-Cunha A, Silva BMD, Porto G, Lopes C. Expression of iron-related proteins in feline and canine mammary gland reveals unexpected accumulation of iron. Biotech Histochem 2017; 92:584-594. [PMID: 29172705 DOI: 10.1080/10520295.2017.1369160] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Dysregulation of cellular iron homeostasis in human breast cancer is reflected by the altered expression of regulatory proteins. The expressions of iron-related proteins in the mammary glands of cats and dogs have not been assessed. We evaluated the expressions of ferritin, ferroportin, hepcidin and transferrin receptor 1 in benign and malignant mammary gland lesions in cats and dogs. Iron deposition was detected using Perls' Prussian blue staining. We found no major differences in the expression of iron-related proteins between benign and malignant mammary gland lesions in either cats or dogs; however, these species exhibited accumulation of iron in benign lesions. Our findings provide an explanation for the absence of higher iron requirements by tumor cells in these animals. Further investigation of local iron homeostasis in cats and dogs and differences in their physiology compared to human breast cancer is required.
Collapse
Affiliation(s)
- O Marques
- a Unit for Multidisciplinary Biomedical Research (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto , Porto.,b Pathology and Molecular Immunology Department , Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto , Porto.,c Basic and Clinical Research on Iron Biology, Molecular and Cell Biology Institute (IBMC) University of Porto , Porto.,d Institute for Research and Innovation in Health Sciences (i3S), University of Porto , Porto
| | - A Canadas
- b Pathology and Molecular Immunology Department , Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto , Porto
| | - F Faria
- b Pathology and Molecular Immunology Department , Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto , Porto
| | - E Oliveira
- a Unit for Multidisciplinary Biomedical Research (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto , Porto.,e Laboratory of Cell Biology, Department of Microscopy , Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto , Porto
| | - I Amorim
- b Pathology and Molecular Immunology Department , Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto , Porto.,d Institute for Research and Innovation in Health Sciences (i3S), University of Porto , Porto.,f Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), University of Porto , Porto
| | - F Seixas
- g Centre of Animal and Veterinary Sciences, University of Trás-os-Montes e Alto Douro (CECAV-UTAD) , Vila Real
| | - A Gama
- g Centre of Animal and Veterinary Sciences, University of Trás-os-Montes e Alto Douro (CECAV-UTAD) , Vila Real
| | - A Lobo-da-Cunha
- a Unit for Multidisciplinary Biomedical Research (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto , Porto.,e Laboratory of Cell Biology, Department of Microscopy , Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto , Porto
| | - B Martins da Silva
- a Unit for Multidisciplinary Biomedical Research (UMIB), Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto , Porto.,b Pathology and Molecular Immunology Department , Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto , Porto
| | - G Porto
- b Pathology and Molecular Immunology Department , Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto , Porto.,c Basic and Clinical Research on Iron Biology, Molecular and Cell Biology Institute (IBMC) University of Porto , Porto.,d Institute for Research and Innovation in Health Sciences (i3S), University of Porto , Porto.,e Laboratory of Cell Biology, Department of Microscopy , Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto , Porto.,f Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), University of Porto , Porto.,g Centre of Animal and Veterinary Sciences, University of Trás-os-Montes e Alto Douro (CECAV-UTAD) , Vila Real.,h Hematology Service, Santo António Hospital, Porto Hospital Centre
| | - C Lopes
- b Pathology and Molecular Immunology Department , Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto , Porto.,i Department of Pathology , Portuguese Oncology Institute (IPO) , Porto , Portugal
| |
Collapse
|
16
|
Schachtschneider KM, Schwind RM, Newson J, Kinachtchouk N, Rizko M, Mendoza-Elias N, Grippo P, Principe DR, Park A, Overgaard NH, Jungersen G, Garcia KD, Maker AV, Rund LA, Ozer H, Gaba RC, Schook LB. The Oncopig Cancer Model: An Innovative Large Animal Translational Oncology Platform. Front Oncol 2017; 7:190. [PMID: 28879168 PMCID: PMC5572387 DOI: 10.3389/fonc.2017.00190] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 08/10/2017] [Indexed: 12/20/2022] Open
Abstract
Despite an improved understanding of cancer molecular biology, immune landscapes, and advancements in cytotoxic, biologic, and immunologic anti-cancer therapeutics, cancer remains a leading cause of death worldwide. More than 8.2 million deaths were attributed to cancer in 2012, and it is anticipated that cancer incidence will continue to rise, with 19.3 million cases expected by 2025. The development and investigation of new diagnostic modalities and innovative therapeutic tools is critical for reducing the global cancer burden. Toward this end, transitional animal models serve a crucial role in bridging the gap between fundamental diagnostic and therapeutic discoveries and human clinical trials. Such animal models offer insights into all aspects of the basic science-clinical translational cancer research continuum (screening, detection, oncogenesis, tumor biology, immunogenicity, therapeutics, and outcomes). To date, however, cancer research progress has been markedly hampered by lack of a genotypically, anatomically, and physiologically relevant large animal model. Without progressive cancer models, discoveries are hindered and cures are improbable. Herein, we describe a transgenic porcine model—the Oncopig Cancer Model (OCM)—as a next-generation large animal platform for the study of hematologic and solid tumor oncology. With mutations in key tumor suppressor and oncogenes, TP53R167H and KRASG12D, the OCM recapitulates transcriptional hallmarks of human disease while also exhibiting clinically relevant histologic and genotypic tumor phenotypes. Moreover, as obesity rates increase across the global population, cancer patients commonly present clinically with multiple comorbid conditions. Due to the effects of these comorbidities on patient management, therapeutic strategies, and clinical outcomes, an ideal animal model should develop cancer on the background of representative comorbid conditions (tumor macro- and microenvironments). As observed in clinical practice, liver cirrhosis frequently precedes development of primary liver cancer or hepatocellular carcinoma. The OCM has the capacity to develop tumors in combination with such relevant comorbidities. Furthermore, studies on the tumor microenvironment demonstrate similarities between OCM and human cancer genomic landscapes. This review highlights the potential of this and other large animal platforms as transitional models to bridge the gap between basic research and clinical practice.
Collapse
Affiliation(s)
| | - Regina M Schwind
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, United States
| | | | | | - Mark Rizko
- College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Nasya Mendoza-Elias
- College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Paul Grippo
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Daniel R Principe
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Alex Park
- College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Nana H Overgaard
- Division of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Gregers Jungersen
- Division of Immunology and Vaccinology, National Veterinary Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Kelly D Garcia
- Biologic Resources Laboratory, University of Illinois at Chicago, Chicago, IL, United States
| | - Ajay V Maker
- Department of Surgical Oncology, University of Illinois at Chicago, Chicago, IL, United States
| | - Laurie A Rund
- Department of Animal Sciences, University of Illinois, Urbana, IL, United States
| | - Howard Ozer
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Ron C Gaba
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, United States
| | - Lawrence B Schook
- Department of Radiology, University of Illinois at Chicago, Chicago, IL, United States.,Department of Animal Sciences, University of Illinois, Urbana, IL, United States
| |
Collapse
|
17
|
Jiao Y, Wang Y, Guo S, Wang G. Glutathione peroxidases as oncotargets. Oncotarget 2017; 8:80093-80102. [PMID: 29108391 PMCID: PMC5668124 DOI: 10.18632/oncotarget.20278] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 06/20/2017] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress is a disturbance in the equilibrium among free radicals, reactive oxygen species, and endogenous antioxidant defense mechanisms. Oxidative stress is a result of imbalance between the production of reactive oxygen and the biological system's ability to detoxify the reactive intermediates or to repair the resulting damage. Mounting evidence has implicated oxidative stress in various physiological and pathological processes, including DNA damage, proliferation, cell adhesion, and survival of cancer cells. Glutathione peroxidases (GPxs) (EC 1.11.1.9) are an enzyme family with peroxidase activity whose main biological roles are to protect organisms from oxidative damage by reducing lipid hydroperoxides as well as free hydrogen peroxide. Currently, 8 sub-members of GPxs have been identified in humans, all capable of reducing H2O2 and soluble fatty acid hydroperoxides. A large number of publications has demonstrated that GPxs have significant roles in different stages of carcinogenesis. In this review, we will update recent progress in the study of the roles of GPxs in cancer. Better mechanistic understanding of GPxs will potentially contribute to the development and advancement of improved cancer treatment models.
Collapse
Affiliation(s)
- Yang Jiao
- Department of Stomatology, PLA Army General Hospital, Beijing, P.R. China
| | - Yirong Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Operative Dentistry and Endodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, P.R. China
| | - Shanchun Guo
- RCMI Cancer Research Center and Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, USA
| | - Guangdi Wang
- RCMI Cancer Research Center and Department of Chemistry, Xavier University of Louisiana, New Orleans, LA, USA
| |
Collapse
|
18
|
Pang LY, Saunders L, Argyle DJ. Epidermal growth factor receptor activity is elevated in glioma cancer stem cells and is required to maintain chemotherapy and radiation resistance. Oncotarget 2017; 8:72494-72512. [PMID: 29069805 PMCID: PMC5641148 DOI: 10.18632/oncotarget.19868] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 07/06/2017] [Indexed: 12/26/2022] Open
Abstract
Glioblastoma remains among the most aggressive of all human and canine malignancies, displaying high mortality rates and limited treatment options. We propose that given the similarities between canine and human gliomas, such as incidence of occurrence, histopathology, molecular characteristics, and response to therapy, that canine gliomas are a natural model of the human disease. A range of human and canine tumours have been shown to harbor specific subpopulations of cells with stem cell-like properties that initiate and maintain neoplasticity while resisting conventional therapies. Here, we show that both canine and human glioma cell lines contain a small population of cancer stem cells (CSCs), and by molecular profiling highlight the important role of the epidermal growth factor receptor (EGFR) pathway in canine CSCs. EGFR signaling is crucial in the regulation of cancer cell proliferation, migration and survival. To date EGFR-targeted interventions alone have been largely ineffective. Our findings confirm that specifically inhibiting EGFR signaling alone has no significant effect on the viability of CSCs. However inhibition of EGFR did enhance the chemo- and radio-sensitivity of both canine and human glioma CSCs, enabling this resistant, tumourigenic population of cells to be effectively targeted by conventional therapies.
Collapse
Affiliation(s)
- Lisa Y Pang
- Royal (Dick) School of Veterinary Studies and Roslin Institute, The University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, Scotland
| | - Lauren Saunders
- Royal (Dick) School of Veterinary Studies and Roslin Institute, The University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, Scotland
| | - David J Argyle
- Royal (Dick) School of Veterinary Studies and Roslin Institute, The University of Edinburgh, Easter Bush, Midlothian, EH25 9RG, Scotland
| |
Collapse
|
19
|
Candelaria PV, Rampoldi A, Harbuzariu A, Gonzalez-Perez RR. Leptin signaling and cancer chemoresistance: Perspectives. World J Clin Oncol 2017; 8:106-119. [PMID: 28439492 PMCID: PMC5385432 DOI: 10.5306/wjco.v8.i2.106] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/20/2016] [Accepted: 03/02/2017] [Indexed: 02/06/2023] Open
Abstract
Obesity is a major health problem and currently is endemic around the world. Obesity is a risk factor for several different types of cancer, significantly promoting cancer incidence, progression, poor prognosis and resistance to anti-cancer therapies. The study of this resistance is critical as development of chemoresistance is a serious drawback for the successful and effective drug-based treatments of cancer. There is increasing evidence that augmented adiposity can impact on chemotherapeutic treatment of cancer and the development of resistance to these treatments, particularly through one of its signature mediators, the adipokine leptin. Leptin is a pro-inflammatory, pro-angiogenic and pro-tumorigenic adipokine that has been implicated in many cancers promoting processes such as angiogenesis, metastasis, tumorigenesis and survival/resistance to apoptosis. Several possible mechanisms that could potentially be developed by cancer cells to elicit drug resistance have been suggested in the literature. Here, we summarize and discuss the current state of the literature on the role of obesity and leptin on chemoresistance, particularly as it relates to breast and pancreatic cancers. We focus on the role of leptin and its significance in possibly driving these proposed chemoresistance mechanisms, and examine its effects on cancer cell survival signals and expansion of the cancer stem cell sub-populations.
Collapse
|
20
|
Yan Y, Wang Y, Zhao P, Ma W, Hu Z, Zhang K. BMI-1 Promotes Self-Renewal of Radio- and Temozolomide (TMZ)-Resistant Breast Cancer Cells. Reprod Sci 2017; 24:1620-1629. [DOI: 10.1177/1933719117697255] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Yanfang Yan
- Department of Gland Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Ying Wang
- Department of Gland Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Pengxin Zhao
- Department of Gland Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Weiyuan Ma
- Department of Gland Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Zhigang Hu
- Department of Gland Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Kaili Zhang
- Department of Gland Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
21
|
Oncogenic roles and drug target of CXCR4/CXCL12 axis in lung cancer and cancer stem cell. Tumour Biol 2016; 37:8515-28. [PMID: 27079871 DOI: 10.1007/s13277-016-5016-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/18/2016] [Indexed: 12/12/2022] Open
Abstract
Although the great progress has been made in diagnosis and therapeutic in lung cancer, it induces the most cancer death worldwide in both males and females. Chemokines, which have chemotactic abilities, contain up to 50 family members. By binding to G protein-coupled receptors (GPCR), holding seven-transmembrane domain, they function in immune cell trafficking and regulation of cell proliferation, differentiation, activation, and migration, homing under both physiologic and pathologic conditions. The alpha-chemokine receptor CXCR4 for the alpha-chemokine stromal cell-derived-factor-1 (SDF-1) is most widely expressed by tumors. In addition to human tissues of the bone marrow, liver, adrenal glands, and brain, the CXC chemokine SDF-1 or CXCL12 is also highly expressed in lung cancer tissues and is associated with lung metastasis. Lung cancer cells have the capabilities to utilize and manipulate the CXCL12/CXCR system to benefit growth and distant spread. CXCL12/CXCR4 axis is a major culprit for lung cancer and has a crucial role in lung cancer initiation and progression by activating cancer stem cell. This review provides an evaluation of CXCL12/CXCR4 as the potential therapeutic target for lung cancers; it also focuses on the synergistic effects of inhibition of CXCL12/CXCR4 axis and immunotherapy as well as chemotherapy. Together, CXCL12/CXCR4 axis can be a potential therapeutic target for lung cancers and has additive effects with immunotherapy.
Collapse
|
22
|
Pang LY, Argyle DJ. Veterinary oncology: Biology, big data and precision medicine. Vet J 2016; 213:38-45. [PMID: 27240913 DOI: 10.1016/j.tvjl.2016.03.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 03/08/2016] [Indexed: 01/28/2023]
Abstract
Despite significant advances in both the understanding and the treatment of cancer, the disease remains one of high mortality and morbidity causes in all species. Increases in survival times in human cancer have increased significantly in the past 25 years but most of these increases have been through small incremental changes. For some cancers, e.g. pancreatic cancer, survival times have not increased significantly in over 100 years. In veterinary oncology, there have been major shifts in the management of cancer in companion animals. Increased availability of specialist centres, coupled with changing attitudes in owners and veterinarians, have meant improvements in veterinary cancer care borne from market pressures and increased awareness and understanding. In this review the changing face of cancer biology over the past 25 years will be examined, and the barriers to clinical progress in veterinary medicine considered. Finally, an optimistic view of the future will be presented with the prospect of greater control over this devastating disease.
Collapse
Affiliation(s)
- Lisa Y Pang
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, Easter Bush, Roslin, Midlothian EH25 9RG, UK
| | - David J Argyle
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, Easter Bush, Roslin, Midlothian EH25 9RG, UK.
| |
Collapse
|
23
|
Spontaneously Arising Canine Glioma as a Potential Model for Human Glioma. J Comp Pathol 2016; 154:169-79. [PMID: 26804204 DOI: 10.1016/j.jcpa.2015.12.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 11/21/2015] [Accepted: 12/01/2015] [Indexed: 01/01/2023]
Abstract
Human gliomas are malignant brain tumours that carry a poor prognosis and are composed of a heterogeneous population of cells. There is a paucity of animal models available for study of these tumours and most have been created by genetic modification. Spontaneously arising canine gliomas may provide a model for the characterization of the human tumours. The present study shows that canine gliomas form a range of immunohistochemical patterns that are similar to those described for human gliomas. The in-vitro sphere assay was used to analyze the expansion and differentiation potential of glioma cells taken from the periphery and centre of canine tumours. Samples from the subventricular zone (SVZ) and contralateral parenchyma were used as positive and negative controls, respectively. The expansion potential for all of these samples was low and cells from only three cultures were expanded for six passages. These three cultures were derived from high-grade gliomas and the cells had been cryopreserved. Most of the cells obtained from the centre of the tumours formed spheres and were expanded, in contrast to samples taken from the periphery of the tumours. Spheres were also formed and expanded from two areas of apparently unaffected brain parenchyma. The neurogenic SVZ contralateral samples also contained progenitor proliferating cells, since all of them were expanded for three to five passages. Differentiation analysis showed that all cultured spheres were multipotential and able to differentiate towards both neurons and glial cells. Spontaneously arising canine gliomas might therefore constitute an animal model for further characterization of these tumours.
Collapse
|
24
|
Supsavhad W, Dirksen WP, Martin CK, Rosol TJ. Animal models of head and neck squamous cell carcinoma. Vet J 2015; 210:7-16. [PMID: 26965084 DOI: 10.1016/j.tvjl.2015.11.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 11/02/2015] [Accepted: 11/11/2015] [Indexed: 12/18/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the most common oral cancer worldwide. Local bone invasion into the maxilla or mandible and metastasis to regional lymph nodes often result in a poor prognosis, decreased quality of life and shortened survival time for HNSCC patients. Poor response to treatment and clinical outcomes are the major concerns in this aggressive cancer. Multiple animal models have been developed to replicate spontaneous HNSCC and investigate genetic alterations and novel therapeutic targets. This review provides an overview of HNSCC as well as the traditional animal models used in HNSCC preclinical research. The value and challenges of each in vivo model are discussed. Similarity between HNSCC in humans and cats and the possibility of using spontaneous feline oral squamous cell carcinoma (FOSCC) as a model for HNSCC in translational research are highlighted.
Collapse
Affiliation(s)
- Wachiraphan Supsavhad
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210, USA
| | - Wessel P Dirksen
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210, USA
| | - Chelsea K Martin
- Department of Pathology and Microbiology, Atlantic Veterinary College, University of Prince Edward Island, 550 University Avenue, Charlottetown, Prince Edward Island C1A 4P3, Canada
| | - Thomas J Rosol
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, 1925 Coffey Road, Columbus, OH 43210, USA.
| |
Collapse
|
25
|
You Q, Guo H, Xu D. Distinct prognostic values and potential drug targets of ALDH1 isoenzymes in non-small-cell lung cancer. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:5087-97. [PMID: 26366059 PMCID: PMC4562757 DOI: 10.2147/dddt.s87197] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Increased aldehyde dehydrogenase 1 (ALDH1) activity has been found in the stem cell populations of leukemia and some solid tumors including non-small-cell lung cancer (NSCLC). However, which ALDH1’s isoenzymes are contributing to ALDH1 activity remains elusive. In addition, the prognostic value of individual ALDH1 isoenzyme is not clear. In the current study, we investigated the prognostic value of ALDH1 isoenzymes in NSCLC patients through the Kaplan–Meier plotter database, which contains updated gene expression data and survival information from a total of 1,926 NSCLC patients. High expression of ALDH1A1 mRNA was found to be correlated to a better overall survival (OS) in all NSCLC patients followed for 20 years (hazard ratio [HR] 0.88 [0.77–0.99], P=0.039). In addition, high expression of ALDH1A1 mRNA was also found to be correlated to better OS in adenocarcinoma (Ade) patients (HR 0.71 [0.57–0.9], P=0.0044) but not in squamous cell carcinoma (SCC) patients (HR 0.92 [0.72–1.16], P=0.48). High expression of ALDH1A2 and ALDH1B1 mRNA was found to be correlated to worser OS in all NSCLC patients, as well as in Ade, but not in SCC patients. High expression of both ALDH1A3 and ALDH1L1 mRNA was not found to be correlated to OS in all NSCLC patients. These results strongly support that ALDH1A1 mRNA in NSCLC is associated with better prognosis. In addition, our current study also supports that ALDH1A2 and ALDH1B1 might be major contributors to the ALDH1 activity in NSCLC, since high expression of ALDH1A2 and ALDH1B1 mRNA was found to be significantly correlated to worser OS in all NSCLC patients. Based on our study, ALDH1A2 and ALDH1B1 might be excellent potential drug targets for NSCLC patients.
Collapse
Affiliation(s)
- Qinghua You
- Department of Pathology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, People's Republic of China
| | - Huanchen Guo
- Department of Respiratory Medicine, Shouguang Hospital of Traditional Chinese Medicine, Shouguang, People's Republic of China
| | - Dongxiang Xu
- Department of Endocrinology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, People's Republic of China
| |
Collapse
|
26
|
Barbieri F, Thellung S, Ratto A, Carra E, Marini V, Fucile C, Bajetto A, Pattarozzi A, Würth R, Gatti M, Campanella C, Vito G, Mattioli F, Pagano A, Daga A, Ferrari A, Florio T. In vitro and in vivo antiproliferative activity of metformin on stem-like cells isolated from spontaneous canine mammary carcinomas: translational implications for human tumors. BMC Cancer 2015; 15:228. [PMID: 25884842 PMCID: PMC4397725 DOI: 10.1186/s12885-015-1235-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 03/20/2015] [Indexed: 12/21/2022] Open
Abstract
Background Cancer stem cells (CSCs) are considered the cell subpopulation responsible for breast cancer (BC) initiation, growth, and relapse. CSCs are identified as self-renewing and tumor-initiating cells, conferring resistance to chemo- and radio-therapy to several neoplasias. Nowadays, th (about 10mM)e pharmacological targeting of CSCs is considered an ineludible therapeutic goal. The antidiabetic drug metformin was reported to suppress in vitro and in vivo CSC survival in different tumors and, in particular, in BC preclinical models. However, few studies are available on primary CSC cultures derived from human postsurgical BC samples, likely because of the limited amount of tissue available after surgery. In this context, comparative oncology is acquiring a relevant role in cancer research, allowing the analysis of larger samples from spontaneous pet tumors that represent optimal models for human cancer. Methods Isolation of primary canine mammary carcinoma (CMC) cells and enrichment in stem-like cell was carried out from fresh tumor specimens by culturing cells in stem-permissive conditions. Phenotypic and functional characterization of CMC-derived stem cells was performed in vitro, by assessment of self-renewal, long-lasting proliferation, marker expression, and drug sensitivity, and in vivo, by tumorigenicity experiments. Corresponding cultures of differentiated CMC cells were used as internal reference. Metformin efficacy on CMC stem cell viability was analyzed both in vitro and in vivo. Results We identified a subpopulation of CMC cells showing human breast CSC features, including expression of specific markers (i.e. CD44, CXCR4), growth as mammospheres, and tumor-initiation in mice. These cells show resistance to doxorubicin but were highly sensitive to metformin in vitro. Finally, in vivo metformin administration significantly impaired CMC growth in NOD-SCID mice, associated with a significant depletion of CSCs. Conclusions Similarly to the human counterpart, CMCs contain stem-like subpopulations representing, in a comparative oncology context, a valuable translational model for human BC, and, in particular, to predict the efficacy of antitumor drugs. Moreover, metformin represents a potential CSC-selective drug for BC, as effective (neo-)adjuvant therapy to eradicate CSC in mammary carcinomas of humans and animals. Electronic supplementary material The online version of this article (doi:10.1186/s12885-015-1235-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Federica Barbieri
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy. .,Centro di Eccellenza per la Ricerca Biomedica (CEBR), University of Genova, Genoa, Italy.
| | - Stefano Thellung
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy. .,Centro di Eccellenza per la Ricerca Biomedica (CEBR), University of Genova, Genoa, Italy.
| | - Alessandra Ratto
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, and National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Genoa, Italy.
| | - Elisa Carra
- Dipartimento di Medicina Sperimentale, University of Genova, Genoa, Italy.
| | - Valeria Marini
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy.
| | - Carmen Fucile
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy.
| | - Adriana Bajetto
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy.
| | - Alessandra Pattarozzi
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy.
| | - Roberto Würth
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy.
| | - Monica Gatti
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy.
| | - Chiara Campanella
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, and National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Genoa, Italy.
| | - Guendalina Vito
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, and National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Genoa, Italy.
| | - Francesca Mattioli
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy.
| | - Aldo Pagano
- Dipartimento di Medicina Sperimentale, University of Genova, Genoa, Italy. .,IRCCS AOU San Martino - IST, Genoa, Italy.
| | | | - Angelo Ferrari
- Istituto Zooprofilattico Sperimentale del Piemonte, Liguria e Valle D'Aosta, and National Reference Center of Veterinary and Comparative Oncology (CEROVEC), Genoa, Italy.
| | - Tullio Florio
- Dipartimento di Medicina Interna, Sezione di Farmacologia, University of Genova, Genoa, Italy. .,Centro di Eccellenza per la Ricerca Biomedica (CEBR), University of Genova, Genoa, Italy.
| |
Collapse
|
27
|
Wu S, Xue W, Huang X, Yu X, Luo M, Huang Y, Liu Y, Bi Z, Qiu X, Bai S. Distinct prognostic values of ALDH1 isoenzymes in breast cancer. Tumour Biol 2015; 36:2421-6. [PMID: 25582316 DOI: 10.1007/s13277-014-2852-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 11/13/2014] [Indexed: 12/31/2022] Open
Abstract
Aldehyde dehydrogenase 1 (ALDH1), also known as aldehyde dehydrogenase 1 family, is composed of six enzymes that are expressed at high levels in stem cells and are involved in the regulation of stem cell function. Increased ALDH1 activity has been found in the stem cell populations of leukemia and some solid tumors including breast cancer (BC). However, which ALDH1's isoenzymes are contributing to ALDH1 activity has not been determined. In addition, the prognostic value of individual ALDH1 isoenzyme is not clear. In the current study, we investigated the prognostic value of ALDH1 isoenzymes in BC patients through "the Kaplan-Meier plotter" (KM plotter) database, in which updated gene expression data and survival information are from a total of 3455 BC patients. ALDH1A1 messenger RNA (mRNA) high expression was found to be correlated to worsen overall survival (OS) for all BC patients. ALDH1A2 and ALDH1L1 mRNA high expressions were found to be correlated to better OS for all BC patients. Both of ALDH1A3 and ALDH1B1 mRNA high expressions were not found to be correlated to OS for all BC patients. These results strongly support that ALDH1A1 was only a biomarker for predicting poor survival of BC patients among ALDH1 isoenzymes. ALDH1A1 might be a major contributor of ALDH1 activity in BC, since only ALDH1A1 mRNA high expression was found to be significantly correlated to worsen OS for all BC patients.
Collapse
Affiliation(s)
- Shaokun Wu
- Department of Oncology, SunYat-Sen Memorial Hospital, SunYat-Sen University, Guangzhou, 510120, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Comparative pathology of canine soft tissue sarcomas: possible models of human non-rhabdomyosarcoma soft tissue sarcomas. J Comp Pathol 2014; 152:22-7. [PMID: 25435513 DOI: 10.1016/j.jcpa.2014.09.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 09/30/2014] [Indexed: 01/06/2023]
Abstract
Comparative analyses of canine and human soft tissue sarcomas (STSs) are lacking. This study compared the histological and immunohistochemical (labelling for desmin, smooth muscle actin [SMA], CD31, pancytokeratin, S100 and CD34) appearance of 32 archived, formalin-fixed, paraffin wax-embedded canine STS tumour specimens by board-certified veterinary and medical pathologists, both blinded to the other's interpretations. Comparison between the veterinary and human diagnoses revealed a generally consistent pattern of interpretation with few notable variations. Most tumours (13/32) were judged to display similar histomorphological appearance to human low-grade spindle cell sarcomas, appearing non-distinctive and morphologically of a fibroblastic/myofibroblastic type. Five canine cases resembled human liposarcoma, but with atypical desmin-positive epithelioid cells present. Five canine cases resembled human spindle cell sarcoma with myxoid features and two additional cases resembled human myxofibrosarcoma. Seven canine cases were noted to resemble human undifferentiated sarcoma. Findings in the present study demonstrate that canine STSs display histological and immunohistochemical features similar to their human equivalents. Because of these cross-species similarities, a particular opportunity exists to understand the biology and treatment of human STS by potentially including dogs as clinical models.
Collapse
|
29
|
Pang LY, Argyle SA, Kamida A, Morrison KO, Argyle DJ. The long-acting COX-2 inhibitor mavacoxib (Trocoxil™) has anti-proliferative and pro-apoptotic effects on canine cancer cell lines and cancer stem cells in vitro. BMC Vet Res 2014; 10:184. [PMID: 25190452 PMCID: PMC4172958 DOI: 10.1186/s12917-014-0184-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Accepted: 08/07/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The NSAID mavacoxib (Trocoxcil™) is a recently described selective COX-2 inhibitor used for the management of inflammatory disease in dogs. It has a long plasma half-life, requiring less frequent dosing and supporting increased owner compliance in treating their dogs. Although the use of NSAIDs has been described in cancer treatment in dogs, there are no studies to date that have examined the utility of mavacoxib specifically. RESULTS In this study we compared the in vitro activity of a short-acting non-selective COX inhibitor (carprofen) with mavacoxib, on cancer cell and cancer stem cell survival. We demonstrate that mavacoxib has a direct cell killing effect on cancer cells, increases apoptosis in cancer cells in a manner that may be independent of caspase activity, and has an inhibitory effect on cell migration. Importantly, we demonstrate that cancer stem cells derived from osteosarcoma cell lines are sensitive to the cytotoxic effect of mavacoxib. CONCLUSIONS Both NSAIDs can inhibit cancer cell proliferation and induce apoptosis in vitro. Importantly, cancer stem cells derived from an osteosarcoma cell line are sensitive to the cytotoxic effect of mavacoxib. Our results suggest that mavacoxib has anti-tumour effects and that this in vitro anti-cancer activity warrants further study.
Collapse
|
30
|
Zhou W, Guo S, Xiong Z, Liu M. Oncogenic role and therapeutic target of transient receptor potential melastatin 7 channel in malignancy. Expert Opin Ther Targets 2014; 18:1177-96. [DOI: 10.1517/14728222.2014.940894] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
31
|
Guth AM, Deogracias M, Dow SW. Comparison of cancer stem cell antigen expression by tumor cell lines and by tumor biopsies from dogs with melanoma and osteosarcoma. Vet Immunol Immunopathol 2014; 161:132-40. [PMID: 25146881 DOI: 10.1016/j.vetimm.2014.07.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 04/25/2014] [Accepted: 07/17/2014] [Indexed: 12/18/2022]
Abstract
Cancer stem cells (CSCs) represent a small subpopulation of tumor cells that play a critical role in initiating and sustaining tumor growth. However, we currently have an incomplete understanding of the expression patterns of CSC antigens in tumors of dogs, nor do we understand how expression of these antigens vary between tumor cell lines and tumor biopsy specimens. Therefore, we used flow cytometry and commonly reported CSC surface and intracellular markers to evaluate the phenotype and overall frequency of CSC subpopulations in tumor cell lines and primary tumor biopsy samples from dogs with melanoma and osteosarcoma. We found that cells expressing common CSC antigens were rare in tumor cell lines, with the exception of tumor cells expressing CD44 and CD90. In contrast, tumor cells expressing conventional CSC antigens such as CD133, CD34, CD44, CD24 and Oct3/4 were much more common in tumor biopsy samples. Notably, the frequency and types of putative CSC subpopulations were very similar in biopsy samples from dogs with either melanoma or osteosarcoma. Our results suggest that the tumor microenvironment significantly influences CSC subpopulations within tumors and that tumor cell lines may not accurately reflect the actual frequency or types of CSC subpopulations present in tumor tissues in vivo.
Collapse
Affiliation(s)
- Amanda M Guth
- Animal Cancer Center, Department of Clinical Sciences, Colorado State University, Ft. Collins, CO 80523, United States
| | - Mike Deogracias
- Animal Cancer Center, Department of Clinical Sciences, Colorado State University, Ft. Collins, CO 80523, United States
| | - Steven W Dow
- Animal Cancer Center, Department of Clinical Sciences, Colorado State University, Ft. Collins, CO 80523, United States.
| |
Collapse
|
32
|
Rasotto R, Goldschmidt MH, Castagnaro M, Carnier P, Caliari D, Zappulli V. The dog as a natural animal model for study of the mammary myoepithelial basal cell lineage and its role in mammary carcinogenesis. J Comp Pathol 2014; 151:166-80. [PMID: 24975897 DOI: 10.1016/j.jcpa.2014.04.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/30/2014] [Accepted: 04/23/2014] [Indexed: 12/31/2022]
Abstract
Basal-like tumours constitute 2-18% of all human breast cancers (HBCs). These tumours have a basal myoepithelial phenotype and it has been hypothesized that they originate from either myoepithelial cells or mammary progenitor cells. They are heterogeneous in morphology, clinical presentation, outcome and response to therapy. Canine mammary carcinomas (CMCs) have epidemiological and biological similarities to HBCs, are frequently biphasic and are composed of two distinct neoplastic populations (epithelial and myoepithelial). The present study evaluates the potential of CMCs as a natural model for basal-like HBCs. Single and double immunohistochemistry was performed on serial sections of 10 normal canine mammary glands and 65 CMCs to evaluate expression of cytokeratin (CK) 8/18, CK5, CK14, α-smooth muscle actin (SMA), calponin (CALP), p63 and vimentin (VIM). The tumours were also evaluated for Ki67 and human epidermal growth factor receptor (HER)-2 expression. A hierarchical model of cell differentiation was established, similar to that for the human breast. We hypothesized that progenitor cells (CK5(+), CK14(+), p63(+) and VIM(+)) differentiate into terminally-differentiated luminal glandular (CK8/18(+)) and myoepithelial (CALP(+), SMA(+) and VIM(+)) cells via intermediary luminal glandular cells (CK5(+), CK14(+) and CK8/CK18(+)) and intermediary myoepithelial cells (CK5(+), CK14(+), p63(+), SMA(+), CALP(+) and VIM(+)). Neoplastic myoepithelial cells in canine complex carcinomas had labelling similar to that of terminally-differentiated myoepithelial cells, while those of carcinomas-and-malignant myoepitheliomas with a more aggressive biological behaviour (i.e. higher frequency of vascular/lymph node invasion and visceral metastases and higher risk of tumour-related death) were comparable with intermediary myoepithelial cells and had significantly higher Ki67 expression. The majority of CMCs examined were negative for expression of HER-2. The biphasic appearance of CMCs with involvement of the myoepithelial component in different stages of cell differentiation may help to define the role of myoepithelial cells in the mammary carcinogenetic process and the heterogeneous nature of basal-like HBCs.
Collapse
Affiliation(s)
- R Rasotto
- Pathology Department, Animal Health Trust, Lanwades Park, Kentford, Newmarket, Suffolk CB8 7UU, UK.
| | - M H Goldschmidt
- Laboratory of Pathology and Toxicology, Department of Pathobiology, University of Pennsylvania, School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - M Castagnaro
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'università 16, 35020 Legnaro, Padova, Italy
| | - P Carnier
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'università 16, 35020 Legnaro, Padova, Italy
| | - D Caliari
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'università 16, 35020 Legnaro, Padova, Italy
| | - V Zappulli
- Department of Comparative Biomedicine and Food Science, University of Padova, Viale dell'università 16, 35020 Legnaro, Padova, Italy
| |
Collapse
|
33
|
Zhou W, Tian Y, Gong H, Guo S, Luo C. Oncogenic role and therapeutic target of leptin signaling in colorectal cancer. Expert Opin Ther Targets 2014; 18:961-71. [PMID: 24946986 DOI: 10.1517/14728222.2014.926889] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Obesity is characterized by high secretion of several cytokines from adipose tissue and is a recognized risk factor for many cancers. Among these cytokines, leptin mainly produced by adipose tissue and cancer cells is the most studied adipokine. Leptin is an activator of cell proliferation, an antiapoptotic molecule and inducer of cancer stem cells in many cell types, and its critical roles in obesity-related tumorigenesis are based on its oncogenic, mitogenic, pro-inflammatory and pro-angiogenic actions. AREAS COVERED These leptin-induced signals and action are critical for their biological effects on energy balance, adiposity, endocrine systems, immunity, angiogenesis as well as oncogenesis. This review focuses on the up-to-date knowledge on the oncogenic role of leptin signaling, clinical significance and specific drug target development in colorectal cancer (CRC). Additionally, leptin-induced angiogenic ability and molecular mechanisms in CRC cells are discussed. EXPERT OPINION Stringent binding affinity of leptin/Ob-R and overexpression of leptin/Ob-R and their targets in cancer cells make it a unique drug target for prevention and treatment of CRC, particularly in obesity colorectal patients.
Collapse
Affiliation(s)
- Weiqiang Zhou
- Shenyang Medical College, Key Laboratory of Environmental Pollution and Microecology of Liaoning Province , No.146 North Huanghe St, Huanggu Dis, Shenyang City, Liaoning Pro 110034 , PR China
| | | | | | | | | |
Collapse
|
34
|
Newman G, Gonzalez-Perez RR. Leptin-cytokine crosstalk in breast cancer. Mol Cell Endocrinol 2014; 382:570-582. [PMID: 23562747 PMCID: PMC3844060 DOI: 10.1016/j.mce.2013.03.025] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2013] [Accepted: 03/26/2013] [Indexed: 02/07/2023]
Abstract
Despite accumulating evidence suggesting a positive correlation between leptin levels, obesity, post-menopause and breast cancer incidence, our current knowledge on the mechanisms involved in these relationships is still incomplete. Since the cloning of leptin in 1994 and its receptor (OB-R) 1 year later by Friedman's laboratory (Zhang et al., 1994) and Tartaglia et al. (Tartaglia et al., 1995), respectively, more than 22,000 papers related to leptin functions in several biological systems have been published (Pubmed, 2012). The ob gene product, leptin, is an important circulating signal for the regulation of body weight. Additionally, leptin plays critical roles in the regulation of glucose homeostasis, reproduction, growth and the immune response. Supporting evidence for leptin roles in cancer has been shown in more than 1000 published papers, with almost 300 papers related to breast cancer (Pubmed, 2012). Specific leptin-induced signaling pathways are involved in the increased levels of inflammatory, mitogenic and pro-angiogenic factors in breast cancer. In obesity, a mild inflammatory condition, deregulated secretion of proinflammatory cytokines and adipokines such as IL-1, IL-6, TNF-α and leptin from adipose tissue, inflammatory and cancer cells could contribute to the onset and progression of cancer. We used an in silico software program, Pathway Studio 9, and found 4587 references citing these various interactions. Functional crosstalk between leptin, IL-1 and Notch signaling (NILCO) found in breast cancer cells could represent the integration of developmental, proinflammatory and pro-angiogenic signals critical for leptin-induced breast cancer cell proliferation/migration, tumor angiogenesis and breast cancer stem cells (BCSCs). Remarkably, the inhibition of leptin signaling via leptin peptide receptor antagonists (LPrAs) significantly reduced the establishment and growth of syngeneic, xenograft and carcinogen-induced breast cancer and, simultaneously decreased the levels of VEGF/VEGFR2, IL-1 and Notch. Inhibition of leptin-cytokine crosstalk might serve as a preventative or adjuvant measure to target breast cancer, particularly in obese women. This review is intended to present an update analysis of leptin actions in breast cancer, highlighting its crosstalk to inflammatory cytokines and growth factors essential for tumor development, angiogenesis and potential role in BCSC.
Collapse
Affiliation(s)
- Gale Newman
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States.
| | - Ruben Rene Gonzalez-Perez
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, United States.
| |
Collapse
|
35
|
Pang LY, Gatenby EL, Kamida A, Whitelaw BA, Hupp TR, Argyle DJ. Global gene expression analysis of canine osteosarcoma stem cells reveals a novel role for COX-2 in tumour initiation. PLoS One 2014; 9:e83144. [PMID: 24416158 PMCID: PMC3885401 DOI: 10.1371/journal.pone.0083144] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 10/30/2013] [Indexed: 12/22/2022] Open
Abstract
Osteosarcoma is the most common primary bone tumour of both children and dogs. It is an aggressive tumour in both species with a rapid clinical course leading ultimately to metastasis. In dogs and children distant metastasis occurs in >80% of individuals treated by surgery alone. Both canine and human osteosarcoma has been shown to contain a sub-population of cancer stem cells (CSCs), which may drive tumour growth, recurrence and metastasis, suggesting that naturally occurring canine osteosarcoma could act as a preclinical model for the human disease. Here we report the successful isolation of CSCs from primary canine osteosarcoma, as well as established cell lines. We show that these cells can form tumourspheres, and demonstrate relative resistance to chemotherapy. We demonstrate similar results for the human osteosarcma cell lines, U2OS and SAOS2. Utilizing the Affymetrix canine microarray, we are able to definitively show that there are significant differences in global gene expression profiles of isolated osteosarcoma stem cells and the daughter adherent cells. We identified 13,221 significant differences (p = 0.05), and significantly, COX-2 was expressed 141-fold more in CSC spheres than daughter adherent cells. To study the role of COX-2 expression in CSCs we utilized the COX-2 inhibitors meloxicam and mavacoxib. We found that COX-2 inhibition had no effect on CSC growth, or resistance to chemotherapy. However inhibition of COX-2 in daughter cells prevented sphere formation, indicating a potential significant role for COX-2 in tumour initiation.
Collapse
Affiliation(s)
- Lisa Y. Pang
- Royal (Dick) School of Veterinary Studies and Roslin Institute, The University of Edinburgh, Easter Bush, Midlothian, United Kingdom
| | - Emma L. Gatenby
- Royal (Dick) School of Veterinary Studies and Roslin Institute, The University of Edinburgh, Easter Bush, Midlothian, United Kingdom
| | - Ayako Kamida
- Royal (Dick) School of Veterinary Studies and Roslin Institute, The University of Edinburgh, Easter Bush, Midlothian, United Kingdom
| | - Bruce A. Whitelaw
- Royal (Dick) School of Veterinary Studies and Roslin Institute, The University of Edinburgh, Easter Bush, Midlothian, United Kingdom
| | - Ted R. Hupp
- Edinburgh Cancer Research UK Centre, The University of Edinburgh, Edinburgh, United Kingdom
| | - David J. Argyle
- Royal (Dick) School of Veterinary Studies and Roslin Institute, The University of Edinburgh, Easter Bush, Midlothian, United Kingdom
| |
Collapse
|
36
|
The application of circulating tumor cells detecting methods in veterinary oncology. Pol J Vet Sci 2013; 16:141-51. [PMID: 23691590 DOI: 10.2478/pjvs-2013-0022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cancers are one of the most common diseases affecting dogs. Many of them develop spontaneously and their biology and histopathology shows many similarities to human cancers. What more, it is proved that there are much more analogies in molecular mechanisms of cancer development between these two species. Human oncology is seeking more and more efficient methods for an early disease detection which results directly in the extended life expectancy of patients affected. One of the most modern trends in the diagnosis of cancer is to detect circulating tumor cells (CTC) in the blood of patients. It is known that these cells are responsible for the formation of metastases in distant organs what results in the patient death. Moreover, it's confirmed that CTC are already present in patients' bloodstream in the early stages of tumor development. There is no doubt that mechanism of metastasis development in dogs is identical and thus the CTC are also present in their bloodstream. Despite the intense researches there is still no optimal method of isolating cancer cells from the blood where they occur extremely rarely. The purpose of this study is to analyze the implications of the detection methods of tumor cells in the blood in veterinary oncology.
Collapse
|
37
|
Denies S, Sanders NN. Recent progress in canine tumor vaccination: potential applications for human tumor vaccines. Expert Rev Vaccines 2013; 11:1375-86. [PMID: 23249236 DOI: 10.1586/erv.12.104] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tumor vaccination holds great promise for the treatment of cancer and research concerning tumor vaccination in dogs is of great interest for veterinary as well as human medicine. Indeed, cancer is the leading cause of death in adult dogs and companion animals are acknowledged as excellent preclinical models for human oncology. The license of the veterinary melanoma vaccine (Oncept™) and Provenge® for the treatment of prostate cancer in men established tumor vaccination as a valid treatment modality for cancer. Although the results with this and other vaccines are promising, there are still some hurdles to overcome. In this article, preclinical and clinical trials with tumor vaccines in dogs are discussed, as well as the surplus value of canine cancer patients for human medicine.
Collapse
Affiliation(s)
- Sofie Denies
- Laboratory of Gene Therapy, Department of Nutrition, Genetics and Ethology, Faculty of Veterinary Medicine, Ghent University, Heidestraat 19, B-9820 Merelbeke, Belgium
| | | |
Collapse
|
38
|
Martin PD, Argyle DJ. Advances in the management of skin cancer. Vet Dermatol 2013; 24:173-80.e38. [PMID: 23331695 DOI: 10.1111/j.1365-3164.2012.01107.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Skin cancer is one of the most commonly diagnosed cancers in the world today in both humans and our pet population. Advances in molecular techniques are now affording us an opportunity to develop therapeutics targeted at specific cancer-related cellular pathways. However, despite progress in conventional treatments, such as chemotherapy and radiation, and the new targeted therapies, some cancers, such as melanoma and cutaneous lymphoma, continue to cause significant mortality and morbidity. This short synopsis is not complete but is aimed at providing an insight into current advanced treatments and horizon therapies for cutaneous malignancies in dogs and cats with comparative aspects.
Collapse
Affiliation(s)
- Pamela D Martin
- Royal (Dick) School of Veterinary Studies and Roslin Institute, Easter Bush, Midlothian EH25 9RG, UK
| | | |
Collapse
|
39
|
Pang LY, Blacking TM, Else RW, Sherman A, Sang HM, Whitelaw BA, Hupp TR, Argyle DJ. Feline mammary carcinoma stem cells are tumorigenic, radioresistant, chemoresistant and defective in activation of the ATM/p53 DNA damage pathway. Vet J 2012; 196:414-23. [PMID: 23219486 PMCID: PMC3696732 DOI: 10.1016/j.tvjl.2012.10.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 10/10/2012] [Accepted: 10/13/2012] [Indexed: 01/16/2023]
Abstract
Cancer stem cells were identified in a feline mammary carcinoma cell line by demonstrating expression of CD133 and utilising the tumour sphere assay. A population of cells was identified that had an invasive, mesenchymal phenotype, expressed markers of pluripotency and enhanced tumour formation in the NOD-SCID mouse and chick embryo models. This population of feline mammary carcinoma stem cells was resistant to chemotherapy and radiation, possibly due to aberrant activation of the ATM/p53 DNA damage pathway. Epithelial–mesenchymal transition was a feature of the invasive phenotype. These data demonstrate that cancer stem cells are a feature of mammary cancer in cats.
Collapse
Affiliation(s)
- L Y Pang
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, Edinburgh EH25 9RG, UK.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Impellizeri JA, Ciliberto G, Aurisicchio L. Electro-gene-transfer as a new tool for cancer immunotherapy in animals. Vet Comp Oncol 2012; 12:310-8. [PMID: 23095099 DOI: 10.1111/vco.12006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Revised: 09/11/2012] [Accepted: 09/20/2012] [Indexed: 12/11/2022]
Abstract
The concept of vaccines based on the direct inoculation of plasmid DNA gained initial proof-of-concept in small rodent species. Further development was hampered by the difficulty to confirm immunogenicity and efficacy in large animal species and, most importantly, in human clinical trials. These negative findings led to the search of complementary technologies which, in combination with intradermal or intramuscular plasmid DNA injection would result in more robust delivery, decreased interindividual variability, clear evidence of clinical efficacy and which would eventually lead to market approval of new vaccine products. The use of high-pressure, needleless devices as an enhancing tool for plasmid DNA delivery led to recent approval by USDA of Oncept™, a therapeutic cancer vaccine directed against tyrosinase for the therapy of melanoma in dogs. An alternative approach to improve plasmid DNA delivery is electro-gene-transfer (EGT). In this article, we briefly review the principles of DNA-EGT and the evidences for efficacy of a telomerase reverse transcriptase vaccine in a dog clinical trial, and provide perspectives for the use of this technology for broader applications in pet animals.
Collapse
Affiliation(s)
- J A Impellizeri
- Department of Oncology, Veterinary Specialty Center of the Hudson Valley, Wappingers Falls, NY, USA
| | | | | |
Collapse
|
41
|
Bentley RT, Mund JA, Pollok KE, Childress MO, Case J. Peripheral blood biomarkers of solid tumor angiogenesis in dogs: a polychromatic flow cytometry pilot study. Vet J 2012; 196:236-40. [PMID: 23063489 DOI: 10.1016/j.tvjl.2012.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Revised: 09/03/2012] [Accepted: 09/04/2012] [Indexed: 12/26/2022]
Abstract
A subset of peripheral blood hematopoietic stem and progenitor cells of bone marrow origin is elevated in humans with solid cancers before treatment and declines with therapy. This biomarker of angiogenesis is not specific to tumor type and has great potential in the objective assessment of treatment response in clinical trials. This pilot study was designed to develop a biomarker of neoangiogenesis in dogs for the diagnosis of cancer, the measurement of treatment response, and the provision of objective data in clinical trials. Polychromatic flow cytometry was used to quantify two subsets of circulating hematopoietic stem and progenitor cells in dogs with spontaneous solid tumors before (n = 8) and after (n = 3) treatment, and normal controls (n = 6). Pro-angiogenic peripheral blood cells of bone marrow origin were detected in all eight cases and the six normal controls; however, there was no statistically significant difference between the two groups. Interestingly, an apparent decline in pro-angiogenic cells was observed after treatment. Bone marrow derived hematopoietic cells appear to contribute to tumor angiogenesis in dogs, as has been previously reported in humans. While the methodology for pro-angiogenic cell quantification in a small number of dogs in the current study did not result in a significant difference from normal controls, an optimized canine polychromatic flow cytometry protocol holds great promise in the development of a canine cancer model and for the objective measurements of treatment response in clinical trials.
Collapse
Affiliation(s)
- R Timothy Bentley
- Department of Veterinary Clinical Sciences, Purdue University, West Lafayette, IN 47905, USA.
| | | | | | | | | |
Collapse
|
42
|
Isolation of stem-like cells from spontaneous feline mammary carcinomas: Phenotypic characterization and tumorigenic potential. Exp Cell Res 2012; 318:847-60. [DOI: 10.1016/j.yexcr.2012.02.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2011] [Revised: 02/06/2012] [Accepted: 02/07/2012] [Indexed: 12/21/2022]
|
43
|
Pang LY, Bergkvist GT, Cervantes-Arias A, Yool DA, Muirhead R, Argyle DJ. Identification of tumour initiating cells in feline head and neck squamous cell carcinoma and evidence for gefitinib induced epithelial to mesenchymal transition. Vet J 2012; 193:46-52. [PMID: 22342216 DOI: 10.1016/j.tvjl.2012.01.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 11/23/2011] [Accepted: 01/07/2012] [Indexed: 01/16/2023]
Abstract
Feline oral squamous cell carcinoma is considered a highly invasive cancer that carries a high level of morbidity. Despite aggressive surgery, patients often succumb to disease, the tumour having inherent insensitivity to radiation and chemotherapy. In this study we sought to identify cells within the feline SCC1 line that have stem cell properties, including inherent resistance mechanisms. When feline cells were subjected to harsh growth conditions, they formed sphere colonies consistent with a stem cell phenotype. Utilising CD133, we were able to identify a small fraction of cells within the population that had enhanced sphere-forming ability, reduced sensitivity to radiation and conventional chemotherapy and demonstrated resistance to the EGFR-targeting drug, gefitinib. In addition, long-term culture of feline SSC1 cells in gefitinib caused a change in cell morphology and gene expression reminiscent of an epithelial to mesenchymal transition. Taken together, these results suggest that feline SCC may be driven by small subset of cancer stem cells.
Collapse
Affiliation(s)
- L Y Pang
- Royal (Dick) School of Veterinary Studies and Roslin Institute, The University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK.
| | | | | | | | | | | |
Collapse
|
44
|
Matos AJF, Baptista CS, Gärtner MF, Rutteman GR. Prognostic studies of canine and feline mammary tumours: the need for standardized procedures. Vet J 2012; 193:24-31. [PMID: 22296767 DOI: 10.1016/j.tvjl.2011.12.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 12/23/2011] [Accepted: 12/31/2011] [Indexed: 10/14/2022]
Abstract
For several years, veterinary oncologists have been struggling with the prognosis of mammary tumours in dogs and cats. Translation of tumour characteristics into prognostic information is an invaluable tool for the use of the most appropriate therapies, as well as for planning innovative therapeutic trials. Moreover, canine and feline spontaneous mammary gland tumours are good models for the study of human breast cancer. Collecting and interpreting information regarding the prognosis of canine and feline mammary tumours is difficult due to the fact that different methods have been applied to study various components and characteristics. This review identifies some of the challenges of prognostic studies of spontaneous canine and feline mammary tumours and suggests standardized procedures to overcome these challenges and facilitate reproducibility and assessment of results.
Collapse
Affiliation(s)
- A J F Matos
- Institute of Biomedical Sciences of Abel Salazar (ICBAS), University of Porto, Portugal.
| | | | | | | |
Collapse
|
45
|
Guo S, Liu M, Wang G, Torroella-Kouri M, Gonzalez-Perez RR. Oncogenic role and therapeutic target of leptin signaling in breast cancer and cancer stem cells. Biochim Biophys Acta Rev Cancer 2012; 1825:207-22. [PMID: 22289780 DOI: 10.1016/j.bbcan.2012.01.002] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2011] [Revised: 01/12/2012] [Accepted: 01/15/2012] [Indexed: 12/17/2022]
Abstract
Significant correlations between obesity and incidence of various cancers have been reported. Obesity, considered a mild inflammatory process, is characterized by a high level of secretion of several cytokines from adipose tissue. These molecules have disparate effects, which could be relevant to cancer development. Among the inflammatory molecules, leptin, mainly produced by adipose tissue and overexpressed with its receptor (Ob-R) in cancer cells is the most studied adipokine. Mutations of leptin or Ob-R genes associated with obesity or cancer are rarely found. However, leptin is an anti-apoptotic molecule in many cell types, and its central roles in obesity-related cancers are based on its pro-angiogenic, pro-inflammatory and mitogenic actions. Notably, these leptin actions are commonly reinforced through entangled crosstalk with multiple oncogenes, cytokines and growth factors. Leptin-induced signals comprise several pathways commonly triggered by many cytokines (i.e., canonical: JAK2/STAT; MAPK/ERK1/2 and PI-3K/AKT1 and, non-canonical signaling pathways: PKC, JNK and p38 MAP kinase). Each of these leptin-induced signals is essential to its biological effects on food intake, energy balance, adiposity, immune and endocrine systems, as well as oncogenesis. This review is mainly focused on the current knowledge of the oncogenic role of leptin in breast cancer. Additionally, leptin pro-angiogenic molecular mechanisms and its potential role in breast cancer stem cells will be reviewed. Strict biunivocal binding-affinity and activation of leptin/Ob-R complex makes it a unique molecular target for prevention and treatment of breast cancer, particularly in obesity contexts.
Collapse
Affiliation(s)
- Shanchun Guo
- Microbiology, Biochemistry & Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA
| | | | | | | | | |
Collapse
|
46
|
Oncogenic role and therapeutic target of leptin signaling in breast cancer and cancer stem cells. BIOCHIMICA ET BIOPHYSICA ACTA 2012. [PMID: 22289780 DOI: 10.1016/j.bbcan.2012.01.002.oncogenic] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Significant correlations between obesity and incidence of various cancers have been reported. Obesity, considered a mild inflammatory process, is characterized by a high level of secretion of several cytokines from adipose tissue. These molecules have disparate effects, which could be relevant to cancer development. Among the inflammatory molecules, leptin, mainly produced by adipose tissue and overexpressed with its receptor (Ob-R) in cancer cells is the most studied adipokine. Mutations of leptin or Ob-R genes associated with obesity or cancer are rarely found. However, leptin is an anti-apoptotic molecule in many cell types, and its central roles in obesity-related cancers are based on its pro-angiogenic, pro-inflammatory and mitogenic actions. Notably, these leptin actions are commonly reinforced through entangled crosstalk with multiple oncogenes, cytokines and growth factors. Leptin-induced signals comprise several pathways commonly triggered by many cytokines (i.e., canonical: JAK2/STAT; MAPK/ERK1/2 and PI-3K/AKT1 and, non-canonical signaling pathways: PKC, JNK and p38 MAP kinase). Each of these leptin-induced signals is essential to its biological effects on food intake, energy balance, adiposity, immune and endocrine systems, as well as oncogenesis. This review is mainly focused on the current knowledge of the oncogenic role of leptin in breast cancer. Additionally, leptin pro-angiogenic molecular mechanisms and its potential role in breast cancer stem cells will be reviewed. Strict biunivocal binding-affinity and activation of leptin/Ob-R complex makes it a unique molecular target for prevention and treatment of breast cancer, particularly in obesity contexts.
Collapse
|
47
|
Canine Mammary Cancer Stem Cells are Radio- and Chemo- Resistant and Exhibit an Epithelial-Mesenchymal Transition Phenotype. Cancers (Basel) 2011; 3:1744-62. [PMID: 24212780 PMCID: PMC3757388 DOI: 10.3390/cancers3021744] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 01/25/2011] [Accepted: 03/22/2011] [Indexed: 12/26/2022] Open
Abstract
Canine mammary carcinoma is the most common cancer among female dogs and is often fatal due to the development of distant metastases. In humans, solid tumors are made up of heterogeneous cell populations, which perform different roles in the tumor economy. A small subset of tumor cells can hold or acquire stem cell characteristics, enabling them to drive tumor growth, recurrence and metastasis. In veterinary medicine, the molecular drivers of canine mammary carcinoma are as yet undefined. Here we report that putative cancer stem cells (CSCs) can be isolated form a canine mammary carcinoma cell line, REM134. We show that these cells have an increased ability to form tumorspheres, a characteristic of stem cells, and that they express embryonic stem cell markers associated with pluripotency. Moreover, canine CSCs are relatively resistant to the cytotoxic effects of common chemotherapeutic drugs and ionizing radiation, indicating that failure of clinical therapy to eradicate canine mammary cancer may be due to the survival of CSCs. The epithelial to mesenchymal transition (EMT) has been associated with cancer invasion, metastasis, and the acquisition of stem cell characteristics. Our results show that canine CSCs predominantly express mesenchymal markers and are more invasive than parental cells, indicating that these cells have a mesenchymal phenotype. Furthermore, we show that canine mammary cancer cells can be induced to undergo EMT by TGFβ and that these cells have an increased ability to form tumorspheres. Our findings indicate that EMT induction can enrich for cells with CSC properties, and provide further insight into canine CSC biology.
Collapse
|
48
|
A vaccine targeting telomerase enhances survival of dogs affected by B-cell lymphoma. Mol Ther 2010; 18:1559-67. [PMID: 20531395 DOI: 10.1038/mt.2010.104] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Canine cancers occur with an incidence similar to that of humans and share many features with human malignancies including histological appearance, tumor genetics, biological behavior, and response to conventional therapies. As observed in humans, the telomerase reverse transcriptase (TERT) activity is largely confined to tumor tissues and absent in the majority of normal dog tissues. Therefore, dog TERT (dTERT) can constitute a valid target for translational cancer immunotherapy. We have evaluated the ability of adenovirus serotype 6 (Ad6) and DNA electroporation (DNA-EP) to induce immune responses against dTERT in dogs affected by malignant lymphoma (ML). The vaccine was combined with standard chemotherapy regimen [cyclophosphamide, vincristine, prednisone (COP)]. dTERT-specific immune response was induced in 13 out of 14 treated animals (93%) and remained detectable and long-lasting with the absence of autoimmunity or other side effects. Most interestingly, the survival time of vaccine/Chemo-treated dogs was significantly increased over historic controls of Chemo-treated animals (>97.8 versus 37 weeks, respectively, P = 0.001). Our results show that Ad6/DNA-EP-based cancer vaccine against dTERT overcomes host immune tolerance, should be combined with chemotherapy, induces long-lasting immune responses, and significantly prolongs the survival of ML canine patients. These data support further evaluation of this approach in human clinical trials.
Collapse
|
49
|
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. [PMID: 19823573 PMCID: PMC2753665 DOI: 10.1371/journal.pmed.1000161] [Citation(s) in RCA: 154] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Chand Khanna and colleagues describe the work of the Comparative Oncology Trials Consortium (COTC), which provides infrastructure and resources to integrate naturally occurring dog cancer models into the development of new human cancer drugs, devices, and imaging techniques.
Collapse
Affiliation(s)
- Ira Gordon
- Radiation Oncology Branch, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Melissa Paoloni
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Christina Mazcko
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Chand Khanna
- Comparative Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|