1
|
Tay KLY, Cowan G, Chatterji S, Conti G, Speirs V. Exploring the One Health Paradigm in Male Breast Cancer. J Mammary Gland Biol Neoplasia 2024; 29:8. [PMID: 38573417 PMCID: PMC10995048 DOI: 10.1007/s10911-024-09560-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 03/15/2024] [Indexed: 04/05/2024] Open
Abstract
How cancer patterns in humans compare to those of other species remains largely unknown and there is an even bigger knowledge gap for rare cancers like male breast cancer. One Health is a convergence of human and animal healthcare that encourages cross-pollination of medical research uniting human and veterinary medicine. Recognising that breast cancer occurs spontaneously in other male species (e.g. primates, canines, felines), and knowing that no laboratory models exist for male breast cancer, which limits our ability to perform functional studies, we explored the feasibility of applying One Health to breast cancer in men by conducting a narrative review of the topic. Spontaneous development of breast cancer was reported in captive male primates and in companion canines and felines. Some parallels in tumour biology of human male breast cancer with canines and primates were found. The age distribution, pattern of biomarker expression and metastasis were similar, with mammary tumours typically detected after two-thirds of average lifespan. However, instances of triple negative and inflammatory breast cancer, which are rarely observed in human male breast cancer, were found in canines and histological classification was inconsistent between species. These disparities need redressing to enable full exploration of the One Health paradigm in rare cancers.
Collapse
Affiliation(s)
- Kirsty Luo-Yng Tay
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - George Cowan
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Subarnarekha Chatterji
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, AB25 2ZD, UK
- Aberdeen Cancer Centre, Aberdeen, UK
| | - Giulia Conti
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, AB25 2ZD, UK
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Valerie Speirs
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, AB25 2ZD, UK.
- Aberdeen Cancer Centre, Aberdeen, UK.
| |
Collapse
|
2
|
Lamouline A, Bersini S, Moretti M. In vitro models of breast cancer bone metastasis: analyzing drug resistance through the lens of the microenvironment. Front Oncol 2023; 13:1135401. [PMID: 37182144 PMCID: PMC10168004 DOI: 10.3389/fonc.2023.1135401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 03/27/2023] [Indexed: 05/16/2023] Open
Abstract
Even though breast cancers usually have a good outcome compared to other tumors, the cancer can progress and create metastases in different parts of the organism, the bone being a predilection locus. These metastases are usually the cause of death, as they are mostly resistant to treatments. This resistance can be caused by intrinsic properties of the tumor, such as its heterogeneity, but it can also be due to the protective role of the microenvironment. By activating signaling pathways protecting cancer cells when exposed to chemotherapy, contributing to their ability to reach dormancy, or even reducing the amount of drug able to reach the metastases, among other mechanisms, the specificities of the bone tissue are being investigated as important players of drug resistance. To this date, most mechanisms of this resistance are yet to be discovered, and many researchers are implementing in vitro models to study the interaction between the tumor cells and their microenvironment. Here, we will review what is known about breast cancer drug resistance in bone metastasis due to the microenvironment and we will use those observations to highlight which features in vitro models should include to properly recapitulate these biological aspects in vitro. We will also detail which elements advanced in vitro models should implement in order to better recapitulate in vivo physiopathology and drug resistance.
Collapse
Affiliation(s)
- Anaïs Lamouline
- Regenerative Medicine Technologies Laboratory, Laboratories for Translational Research (LRT), Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, EOC, Lugano, Switzerland
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milano, Italy
| | - Simone Bersini
- Regenerative Medicine Technologies Laboratory, Laboratories for Translational Research (LRT), Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, EOC, Lugano, Switzerland
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
| | - Matteo Moretti
- Regenerative Medicine Technologies Laboratory, Laboratories for Translational Research (LRT), Ente Ospedaliero Cantonale (EOC), Bellinzona, Switzerland
- Service of Orthopaedics and Traumatology, Department of Surgery, EOC, Lugano, Switzerland
- Euler Institute, Faculty of Biomedical Sciences, Università della Svizzera italiana (USI), Lugano, Switzerland
- Cell and Tissue Engineering Laboratory, IRCCS Istituto Ortopedico Galeazzi, Milano, Italy
| |
Collapse
|
3
|
Yang P, Freeman ZT, Dysko RC, Hoenerhoff MJ. Degenerative Myelopathy and Neuropathy in NOD. Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) Mice Caused by Lactate Dehydrogenase-Elevating Virus (LDV). Toxicol Pathol 2022; 50:390-396. [PMID: 35450478 DOI: 10.1177/01926233221091747] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Following implantation of patient-derived xenograft (PDX) breast carcinomas from three separate individuals, 33/51 female NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mice presented with progressive, unilateral to bilateral, ascending hindlimb paresis to paralysis. Mice were mildly dehydrated, in thin to poor body condition, with reduced to absent hindlimb withdrawal reflex and deep pain sensation. Microscopically, there was variable axonal swelling, vacuolation, and dilation of myelin sheaths within the ventral spinal cord and spinal nerve roots of the thoracolumbar and sacral spinal cord, as well as within corresponding sciatic nerves. Results of PCR screening of PDX samples obtained at necropsy and pooled environmental swabs from the racks housing affected animals were positive for lactate dehydrogenase-elevating virus (LDV). LDV is transmitted through animal-animal contact or commonly as a contaminant of biologic materials of mouse origin. Infection is associated with progressive degenerative myelopathy and neuropathy in strains of mice harboring endogenous retrovirus (AKR, C58), or in immunosuppressed strains (NOD-SCID, Foxn1nu), and can interfere with normal immune responses and alter engraftment and growth of xenograft tumors in immunosuppressed mice. This is the first reported series of LDV-induced poliomyelitis in NSG mice and should be recognized as a potentially significant confounder to biomedical studies utilizing immunodeficient xenograft models.
Collapse
Affiliation(s)
- P Yang
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Z T Freeman
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - R C Dysko
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - M J Hoenerhoff
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, Michigan, USA.,In Vivo Animal Core, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
4
|
RANKL immunisation inhibits prostate cancer metastasis by modulating EMT through a RANKL-dependent pathway. Sci Rep 2021; 11:12186. [PMID: 34108600 PMCID: PMC8190078 DOI: 10.1038/s41598-021-91721-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 05/31/2021] [Indexed: 12/17/2022] Open
Abstract
Prostate cancer (PCa) morbidity in the majority of patients is due to metastatic events, which are a clinical obstacle. Therefore, a better understanding of the mechanism underlying metastasis is imperative if we are to develop novel therapeutic strategies. Receptor activator of nuclear factor kappa-B (NF-κB) ligand (RANKL) regulates bone remodelling. Thus, agents that suppress RANKL signalling may be useful pharmacological treatments. Here, we used preclinical experimental models to investigate whether an inactive form of RANKL affects bone metastasis in RANKL-induced PCa. RANKL was associated with epithelial–mesenchymal transition (EMT) and expression of metastasis-related genes in PC3 cells. Therefore, we proposed a strategy to induce anti-cytokine antibodies using mutant RANKL as an immunogen. RANKL promoted migration and invasion of PC3 cells through EMT, and induced a significant increase in binding of β-catenin to TCF-4, an EMT-induced transcription factor in PCa cells, via mitogen-activated protein kinase and β-catenin/TCF-4 signalling. Thus, RANKL increased EMT and the metastatic properties of PC3 cells, suggesting a role as a therapeutic target to prevent PCa metastasis. Treatment with mutant RANKL reduced EMT and metastasis of PC3 PCa cells in an experimental metastasis model. Thus, mutant RANKL could serve as a potential vaccine to prevent and treat metastatic PCa.
Collapse
|
5
|
Trovatelli M, Bassi J, Giudice C, Ferrari R, Longo M, Brizzola S. Prostatic leiomyosarcoma in a dog treated using a multimodal approach. J Vet Intern Med 2021; 35:1105-1110. [PMID: 33625746 PMCID: PMC7995369 DOI: 10.1111/jvim.16081] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 02/04/2021] [Accepted: 02/04/2021] [Indexed: 11/29/2022] Open
Abstract
Prostatic leiomyosarcoma is an uncommon tumor encountered in male dogs, with only 2 cases reported in the veterinary literature with no follow-up described. A 12-year-old male intact German Wirehaired Pointer presented for evaluation of straining to defecate and urinate. Whole body computed tomography (CT) examination identified a spherical multicavitary expansile mass arising from the prostate gland and severely obliterating the pelvic canal. Partial subcapsular prostatectomy was performed, and histological and immunohistochemical results were consistent with prostatic leiomyosarcoma. Metronomic cyclophosphamide and nonsteroidal anti-inflammatory drugs were administered as adjuvant chemotherapy. Follow-up CT 10 months later indicated no signs of recurrence or metastasis. To the best of our knowledge, this patient represents the first report of successful multidisciplinary treatment consisting of partial subcapsular prostatectomy and adjuvant chemotherapy for prostatic leiomyosarcoma in a dog. After 15 months of follow-up, the patient remained recurrence-free without metastasis.
Collapse
Affiliation(s)
- Marco Trovatelli
- Department of Veterinary Medicine, Università degli Studi di Milano, Lodi, Italy
| | - Jessica Bassi
- Veterinary and Zootechnical Experimental Clinical Centre, Università degli Studi di Milano, Lodi, Italy
| | - Chiara Giudice
- Department of Veterinary Medicine, Università degli Studi di Milano, Lodi, Italy
| | - Roberta Ferrari
- Department of Veterinary Medicine, Università degli Studi di Milano, Lodi, Italy
| | - Maurizio Longo
- Centro Traumatologico Ortopedico Veterinario, Arenzano (GE), Italy
| | - Stefano Brizzola
- Department of Veterinary Medicine, Università degli Studi di Milano, Lodi, Italy
| |
Collapse
|
6
|
Elshafae SM, Dirksen WP, Alasonyalilar-Demirer A, Breitbach J, Yuan S, Kantake N, Supsavhad W, Hassan BB, Attia Z, Rosol TJ. Canine prostatic cancer cell line (LuMa) with osteoblastic bone metastasis. Prostate 2020; 80:698-714. [PMID: 32348616 PMCID: PMC7291846 DOI: 10.1002/pros.23983] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/28/2020] [Accepted: 04/02/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Osteoblastic bone metastasis represents the most common complication in men with prostate cancer (PCa). During progression and bone metastasis, PCa cells acquire properties similar to bone cells in a phenomenon called osteomimicry, which promotes their ability to metastasize, proliferate, and survive in the bone microenvironment. The mechanism of osteomimicry resulting in osteoblastic bone metastasis is unclear. METHODS We developed and characterized a novel canine prostatic cancer cell line (LuMa) that will be useful to investigate the relationship between osteoblastic bone metastasis and osteomimicry in PCa. The LuMa cell line was established from a primary prostate carcinoma of a 13-year old mixed breed castrated male dog. Cell proliferation and gene expression of LuMa were measured and compared to three other canine prostatic cancer cell lines (Probasco, Ace-1, and Leo) in vitro. The effect of LuMa cells on calvaria and murine preosteoblastic (MC3T3-E1) cells was measured by quantitative reverse-transcription polymerase chain reaction and alkaline phosphatase assay. LuMa cells were transduced with luciferase for monitoring in vivo tumor growth and metastasis using different inoculation routes (subcutaneous, intratibial [IT], and intracardiac [IC]). Xenograft tumors and metastases were evaluated using radiography and histopathology. RESULTS After left ventricular injection, LuMa cells metastasized to bone, brain, and adrenal glands. IT injections induced tumors with intramedullary new bone formation. LuMa cells had the highest messenger RNA levels of osteomimicry genes (RUNX2, RANKL, and Osteopontin [OPN]), CD44, E-cadherin, and MYOF compared to Ace-1, Probasco, and Leo cells. LuMa cells induced growth in calvaria defects and modulated gene expression in MC3T3-E1 cells. CONCLUSIONS LuMa is a novel canine PCa cell line with osteomimicry and stemness properties. LuMa cells induced osteoblastic bone formation in vitro and in vivo. LuMa PCa cells will serve as an excellent model for studying the mechanisms of osteomimicry and osteoblastic bone and brain metastasis in prostate cancer.
Collapse
Affiliation(s)
- Said M. Elshafae
- Dept. of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
- Dept. of Pathology, Faculty of Veterinary medicine, Benha University, Benha, Egypt
- Dept. of Neuroscience and Pharmacology, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | - Wessel P. Dirksen
- Dept. of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
- Dept. of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Aylin Alasonyalilar-Demirer
- Dept. of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
- Department of Pathology, Faculty of Veterinary Medicine, Bursa Uludag University, Turkey
| | - Justin Breitbach
- Dept. of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
| | - Shiyu Yuan
- Dept. of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Noriko Kantake
- Dept. of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
| | - Wachiraphan Supsavhad
- Dept. of Pathology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
| | - Bardes B. Hassan
- Dept. of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
- Dept. of Pathology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Zayed Attia
- Dept. of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, OH, USA
- Dept. of Animal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Sadat City University, Sadat City, Egypt
| | - Thomas J. Rosol
- Dept. of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH, USA
- Correspondence to: Dr. Thomas Rosol, Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, 225 Irvine Hall, Athens, OH 45701, USA. , Phone: 740.593.2405
| |
Collapse
|
7
|
Fiordelisi MF, Cavaliere C, Auletta L, Basso L, Salvatore M. Magnetic Resonance Imaging for Translational Research in Oncology. J Clin Med 2019; 8:jcm8111883. [PMID: 31698697 PMCID: PMC6912299 DOI: 10.3390/jcm8111883] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/28/2019] [Accepted: 10/29/2019] [Indexed: 12/19/2022] Open
Abstract
The translation of results from the preclinical to the clinical setting is often anything other than straightforward. Indeed, ideas and even very intriguing results obtained at all levels of preclinical research, i.e., in vitro, on animal models, or even in clinical trials, often require much effort to validate, and sometimes, even useful data are lost or are demonstrated to be inapplicable in the clinic. In vivo, small-animal, preclinical imaging uses almost the same technologies in terms of hardware and software settings as for human patients, and hence, might result in a more rapid translation. In this perspective, magnetic resonance imaging might be the most translatable technique, since only in rare cases does it require the use of contrast agents, and when not, sequences developed in the lab can be readily applied to patients, thanks to their non-invasiveness. The wide range of sequences can give much useful information on the anatomy and pathophysiology of oncologic lesions in different body districts. This review aims to underline the versatility of this imaging technique and its various approaches, reporting the latest preclinical studies on thyroid, breast, and prostate cancers, both on small laboratory animals and on human patients, according to our previous and ongoing research lines.
Collapse
|
8
|
What is the Best Radionuclide for Immuno-PET of Multiple Myeloma? A Comparison Study Between 89Zr- and 64Cu-Labeled Anti-CD138 in a Preclinical Syngeneic Model. Int J Mol Sci 2019; 20:ijms20102564. [PMID: 31137758 PMCID: PMC6567828 DOI: 10.3390/ijms20102564] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 05/13/2019] [Accepted: 05/22/2019] [Indexed: 12/16/2022] Open
Abstract
Although positron emission tomography (PET) imaging with 18-Fluorodeoxyglucose (18F-FDG) is a promising technique in multiple myeloma (MM), the development of other radiopharmaceuticals seems relevant. CD138 is currently used as a standard marker for the identification of myeloma cells and could be used in phenotype tumor imaging. In this study, we used an anti-CD138 murine antibody (9E7.4) radiolabeled with copper-64 (64Cu) or zirconium-89 (89Zr) and compared them in a syngeneic mouse model to select the optimal tracers for MM PET imaging. Then, 9E7.4 was conjugated to TE2A-benzyl isothiocyanate (TE2A) and desferrioxamine (DFO) chelators for 64Cu and 89Zr labeling, respectively. 64Cu-TE2A-9E7.4 and 89Zr-DFO-9E7.4 antibodies were evaluated by PET imaging and biodistribution studies in C57BL/KaLwRij mice bearing either 5T33-MM subcutaneous tumors or bone lesions and were compared to 18F-FDG-PET imaging. In biodistribution and PET studies, 64Cu-TE2A-9E7.4 and 89Zr-DFO-9E7.4 displayed comparable good tumor uptake of subcutaneous tumors. On the bone lesions, PET imaging with 64Cu-TE2A-9E7.4 and 89Zr-DFO-9E7.4 showed higher uptake than with 18F-FDG-PET. Comparison of both 9E7.4 conjugates revealed higher nonspecific bone uptakes of 89Zr-DFO-9E7.4 than 64Cu-TE2A-9E7.4. Because of free 89Zr’s tropism for bone when using 89Zr-anti-CD138, 64Cu-anti-CD138 antibody had the most optimal tumor-to-nontarget tissue ratios for translation into humans as a specific new imaging radiopharmaceutical agent in MM.
Collapse
|
9
|
Kunihiro AG, Brickey JA, Frye JB, Luis PB, Schneider C, Funk JL. Curcumin, but not curcumin-glucuronide, inhibits Smad signaling in TGFβ-dependent bone metastatic breast cancer cells and is enriched in bone compared to other tissues. J Nutr Biochem 2019; 63:150-156. [PMID: 30393127 PMCID: PMC6296872 DOI: 10.1016/j.jnutbio.2018.09.021] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 09/24/2018] [Accepted: 09/27/2018] [Indexed: 01/21/2023]
Abstract
Breast cancer (BCa) bone metastases (BMETs) drive osteolysis via a feed-forward loop involving tumoral secretion of osteolytic factors (e.g., PTHrP) induced by bone-matrix-derived growth factors (e.g., TGFβ). In prior experiments, turmeric-derived curcumin inhibited in vivo BMET progression and in vitro TGFβ/Smad-signaling in a TGFβ-stimulated PTHrP-dependent human xenograft BCa BMET model (MDA-SA cells). However, it is unclear whether curcumin or curcumin-glucuronide mediates in vivo protection since curcumin-glucuronide is the primary circulating metabolite in rodents and in humans. Thus, effects of curcumin vs. curcumin-glucuronide on Smad-dependent TGFβ signaling were compared in a series of BCa cell lines forming TGFβ-dependent BMET in murine models, and tissue-specific metabolism of curcumin in mice was examined by LC-MS. While curcumin inhibited TGFβ-receptor-mediated Smad2/3 phosphorylation in all BCa cells studied (human MDA-SA, MDA-1833, MDA-2287 and murine 4T1 cells), curcumin-glucuronide did not. Similarly, curcumin, but not curcumin-glucuronide, blocked TGFβ-stimulated secretion of PTHrP from MDA-SA and 4T1 cells. Because the predominant serum metabolite, curcumin-glucuronide, lacked bioactivity, we examined tissue-specific metabolism of curcumin in mice. Compared to serum and other organs, free curcumin (both absolute and percentage of total) was significantly increased in bone, which was also a rich source of enzymatic deglucuronidation activity. Thus, curcumin, and not curcumin-glucuronide, appears to inhibit bone-tropic BCa cell TGFβ-signaling and to undergo site-specific activation (deconjugation) within the bone microenvironment. These findings suggest that circulating curcumin-glucuronide may act as a prodrug that preferentially targets bone, a process that may contribute to the bone-protective effects of curcumin and other highly glucuronidated dietary polyphenols.
Collapse
Affiliation(s)
- Andrew G Kunihiro
- Department of Nutritional Sciences, University of Arizona, 1656 E. Mabel St, Medical Research Building Rm 418, Tucson, AZ 85719, USA.
| | - Julia A Brickey
- Department of Medicine, University of Arizona, 1656 E. Mabel St, Medical Research Building Rm 418, Tucson, AZ 85719, USA.
| | - Jennifer B Frye
- Department of Medicine, University of Arizona, 1656 E. Mabel St, Medical Research Building Rm 418, Tucson, AZ 85719, USA.
| | - Paula B Luis
- Department of Pharmacology, Vanderbilt University, 23(rd) Ave S at Pierce RRB, Rm 514, Nashville, TN 37232-6602, USA.
| | - Claus Schneider
- Department of Pharmacology, Vanderbilt University, 23(rd) Ave S at Pierce RRB, Rm 514, Nashville, TN 37232-6602, USA.
| | - Janet L Funk
- Department of Nutritional Sciences, University of Arizona, 1656 E. Mabel St, Medical Research Building Rm 418, Tucson, AZ 85719, USA; Department of Medicine, University of Arizona, 1656 E. Mabel St, Medical Research Building Rm 418, Tucson, AZ 85719, USA.
| |
Collapse
|
10
|
Kumar A, Dhar S, Campanelli G, Butt NA, Schallheim JM, Gomez CR, Levenson AS. MTA1 drives malignant progression and bone metastasis in prostate cancer. Mol Oncol 2018; 12:1596-1607. [PMID: 30027683 PMCID: PMC6120234 DOI: 10.1002/1878-0261.12360] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/05/2018] [Accepted: 07/13/2018] [Indexed: 12/27/2022] Open
Abstract
Prostate cancer often metastasizes to the bone, leading to morbidity and mortality. While metastasis-associated protein 1 (MTA1) is highly overexpressed in metastatic tumors and bone metastatic lesions, its exact role in the development of metastasis is unknown. Here, we report the role of MTA1 in prostate cancer progression and bone metastasis in vitro and in vivo. We found that MTA1 silencing diminished formation of bone metastases and impaired tumor growth in intracardiac and subcutaneous prostate cancer xenografts, respectively. This was attributed to reduced colony formation, invasion, and migration capabilities of MTA1 knockdown cells. Mechanistic studies revealed that MTA1 silencing led to a significant decrease in the expression of cathepsin B (CTSB), a cysteine protease critical for bone metastasis, with an expected increase in the levels of E-cadherin in both cells and xenograft tumors. Moreover, meta-analysis of clinical samples indicated a positive correlation between MTA1 and CTSB. Together, these results demonstrate the critical role of MTA1 as an upstream regulator of CTSB-mediated events associated with cell invasiveness and raise the possibility that targeting MTA1/CTSB signaling in the tumor may prevent the development of bone metastasis in prostate cancer.
Collapse
Affiliation(s)
- Avinash Kumar
- Arnold & Marie Schwartz College of Pharmacy and Health SciencesLong Island UniversityBrooklynNYUSA
- Cancer InstituteUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Swati Dhar
- Cancer InstituteUniversity of Mississippi Medical CenterJacksonMSUSA
- Present address:
Department of PediatricsFeinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Gisella Campanelli
- Arnold & Marie Schwartz College of Pharmacy and Health SciencesLong Island UniversityBrooklynNYUSA
| | - Nasir A. Butt
- Cancer InstituteUniversity of Mississippi Medical CenterJacksonMSUSA
- Department of PathologyUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Jason M. Schallheim
- Department of PathologyUniversity of Mississippi Medical CenterJacksonMSUSA
- Present address:
Anne Arundel Medical CenterAnnapolisMDUSA
| | - Christian R. Gomez
- Cancer InstituteUniversity of Mississippi Medical CenterJacksonMSUSA
- Department of PathologyUniversity of Mississippi Medical CenterJacksonMSUSA
- Department of Radiation OncologyUniversity of Mississippi Medical CenterJacksonMSUSA
| | - Anait S. Levenson
- Arnold & Marie Schwartz College of Pharmacy and Health SciencesLong Island UniversityBrooklynNYUSA
- Cancer InstituteUniversity of Mississippi Medical CenterJacksonMSUSA
- Department of PathologyUniversity of Mississippi Medical CenterJacksonMSUSA
- Present address:
Veterinary MedicineLong Island UniversityBrookvilleNYUSA
| |
Collapse
|
11
|
Sun Q, Choudhary S, Mannion C, Kissin Y, Zilberberg J, Lee WY. Ex vivo construction of human primary 3D-networked osteocytes. Bone 2017; 105:245-252. [PMID: 28942121 PMCID: PMC5690542 DOI: 10.1016/j.bone.2017.09.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 09/06/2017] [Accepted: 09/20/2017] [Indexed: 02/07/2023]
Abstract
A human bone tissue model was developed by constructing ex vivo the 3D network of osteocytes via the biomimetic assembly of primary human osteoblastic cells with 20-25μm microbeads and subsequent microfluidic perfusion culture. The biomimetic assembly: (1) enabled 3D-constructed cells to form cellular network via processes with an average cell-to-cell distance of 20-25μm, and (2) inhibited cell proliferation within the interstitial confine between the microbeads while the confined cells produced extracellular matrix (ECM) to form a mechanically integrated structure. The mature osteocytic expressions of SOST and FGF23 genes became significantly higher, especially for SOST by 250 folds during 3D culture. The results validate that the bone tissue model: (1) consists of 3D cellular network of primary human osteocytes, (2) mitigates the osteoblastic differentiation and proliferation of primary osteoblast-like cells encountered in 2D culture, and (3) therefore reproduces ex vivo the phenotype of human 3D-networked osteocytes. The 3D tissue construction approach is expected to provide a clinically relevant and high-throughput means for evaluating drugs and treatments that target bone diseases with in vitro convenience.
Collapse
Affiliation(s)
- Qiaoling Sun
- Department of Materials Science and Chemical Engineering, Stevens Institute of Technology, Hoboken, NJ, USA
| | - Saba Choudhary
- Department of Biomedical Engineering, Chemistry and Biological Sciences, Stevens Institute of Technology, Hoboken, NJ, USA
| | - Ciaran Mannion
- Department of Pathology, Hackensack University Medical Center, Hackensack, NJ, USA
| | - Yair Kissin
- Hackensack University Medical Center, Hackensack, NJ, USA
| | - Jenny Zilberberg
- Research Department, Hackensack University Medical Center, Hackensack, NJ, USA.
| | - Woo Y Lee
- Department of Materials Science and Chemical Engineering, Stevens Institute of Technology, Hoboken, NJ, USA.
| |
Collapse
|
12
|
Linde N, Fluegen G, Aguirre-Ghiso JA. The Relationship Between Dormant Cancer Cells and Their Microenvironment. Adv Cancer Res 2016; 132:45-71. [PMID: 27613129 PMCID: PMC5342905 DOI: 10.1016/bs.acr.2016.07.002] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The majority of cancer deaths are due to metastases that can occur years or decades after primary tumor diagnosis and treatment. Disseminated tumor cells (DTCs) surviving in a dormant state in target organs appear to explain the timing of this phenomenon. Knowledge on this process is important as it might provide a window of opportunity to prevent recurrences by eradicating dormant DTCs and/or by maintaining DTCs in a dormant state. Importantly, this research might offer markers of dormancy for early monitoring of metastatic relapse. However, our understanding of the mechanisms underlying the regulation of entry into and exit from dormancy is still limited and crippling any therapeutic opportunity. While cancer cell-intrinsic signaling pathways have been linked to dormancy regulation, it is likely that these pathways and the switch controlling reactivation from dormancy are regulated by microenvironmental cues. Here we review and discuss recent findings on how the microenvironment regulates cancer dormancy and raise new questions that may help advance the field.
Collapse
Affiliation(s)
- N Linde
- Tisch Cancer Institute, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY, United States.
| | - G Fluegen
- Tisch Cancer Institute, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY, United States.
| | - J A Aguirre-Ghiso
- Tisch Cancer Institute, Black Family Stem Cell Institute, Mount Sinai School of Medicine, New York, NY, United States.
| |
Collapse
|
13
|
Murine models of breast cancer bone metastasis. BONEKEY REPORTS 2016; 5:804. [PMID: 27867497 DOI: 10.1038/bonekey.2016.31] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 04/02/2016] [Indexed: 01/28/2023]
Abstract
Bone metastases cause significant morbidity and mortality in late-stage breast cancer patients and are currently considered incurable. Investigators rely on translational models to better understand the pathogenesis of skeletal complications of malignancy in order to identify therapeutic targets that may ultimately prevent and treat solid tumor metastasis to bone. Many experimental models of breast cancer bone metastases are in use today, each with its own caveats. In this methods review, we characterize the bone phenotype of commonly utilized human- and murine-derived breast cell lines that elicit osteoblastic and/or osteolytic destruction of bone in mice and report methods for optimizing tumor-take in murine models of bone metastasis. We then provide protocols for four of the most common xenograft and syngeneic inoculation routes for modeling breast cancer metastasis to the skeleton in mice, including the intra-cardiac, intra-arterial, orthotopic and intra-tibial methods of tumor cell injection. Recommendations for in vivo and ex vivo assessment of tumor progression and bone destruction are provided, followed by discussion of the strengths and limitations of the available tools and translational models that aid investigators in the study of breast cancer metastasis to bone.
Collapse
|
14
|
Holen I, Nutter F, Wilkinson JM, Evans CA, Avgoustou P, Ottewell PD. Human breast cancer bone metastasis in vitro and in vivo: a novel 3D model system for studies of tumour cell-bone cell interactions. Clin Exp Metastasis 2015; 32:689-702. [PMID: 26231669 DOI: 10.1007/s10585-015-9737-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 07/28/2015] [Indexed: 01/09/2023]
Abstract
Bone is established as the preferred site of breast cancer metastasis. However, the precise mechanisms responsible for this preference remain unidentified. In order to improve outcome for patients with advanced breast cancer and skeletal involvement, we need to better understand how this process is initiated and regulated. As bone metastasis cannot be easily studied in patients, researchers have to date mainly relied on in vivo xenograft models. A major limitation of these is that they do not contain a human bone microenvironment, increasingly considered to be an important component of metastases. In order to address this shortcoming, we have developed a novel humanised bone model, where 1 × 10(5) luciferase-expressing MDA-MB-231 or T47D human breast tumour cells are seeded on viable human subchaodral bone discs in vitro. These discs contain functional osteoclasts 2-weeks after in vitro culture and positive staining for calcine 1-week after culture demonstrating active bone resorption/formation. In vitro inoculation of MDA-MB-231 or T47D cells colonised human bone cores and remained viable for <4 weeks, however, use of matrigel to enhance adhesion or a moving platform to increase diffusion of nutrients provided no additional advantage. Following colonisation by the tumour cells, bone discs pre-seeded with MDA-MB-231 cells were implanted subcutaneously into NOD SCID mice, and tumour growth monitored using in vivo imaging for up to 6 weeks. Tumour growth progressed in human bone discs in 80 % of the animals mimicking the later stages of human bone metastasis. Immunohistochemical and PCR analysis revealed that growing MDA-MB-231 cells in human bone resulted in these cells acquiring a molecular phenotype previously associated with breast cancer bone metastases. MDA-MB-231 cells grown in human bone discs showed increased expression of IL-1B, HRAS and MMP9 and decreased expression of S100A4, whereas, DKK2 and FN1 were unaltered compared with the same cells grown in mammary fat pads of mice not implanted with human bone discs.
Collapse
Affiliation(s)
- I Holen
- Academic Unit of Clinical Oncology, Department of Oncology, Mellanby Centre for Bone Research, Medical School, University of Sheffield, Sheffield, S10 2RX, UK
| | - F Nutter
- Academic Unit of Clinical Oncology, Department of Oncology, Mellanby Centre for Bone Research, Medical School, University of Sheffield, Sheffield, S10 2RX, UK
| | - J M Wilkinson
- Department of Human Metabolism, Mellanby Centre for Bone Research, Medical School, University of Sheffield, Sheffield, S10 2RX, UK
| | - C A Evans
- Academic Unit of Clinical Oncology, Department of Oncology, Mellanby Centre for Bone Research, Medical School, University of Sheffield, Sheffield, S10 2RX, UK
| | - P Avgoustou
- Academic Unit of Clinical Oncology, Department of Oncology, Mellanby Centre for Bone Research, Medical School, University of Sheffield, Sheffield, S10 2RX, UK
| | - Penelope D Ottewell
- Academic Unit of Clinical Oncology, Department of Oncology, Mellanby Centre for Bone Research, Medical School, University of Sheffield, Sheffield, S10 2RX, UK.
| |
Collapse
|
15
|
Simmons JK, Hildreth BE, Supsavhad W, Elshafae SM, Hassan BB, Dirksen WP, Toribio RE, Rosol TJ. Animal Models of Bone Metastasis. Vet Pathol 2015; 52:827-41. [PMID: 26021553 DOI: 10.1177/0300985815586223] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Bone is one of the most common sites of cancer metastasis in humans and is a significant source of morbidity and mortality. Bone metastases are considered incurable and result in pain, pathologic fracture, and decreased quality of life. Animal models of skeletal metastases are essential to improve the understanding of the molecular pathways of cancer metastasis and growth in bone and to develop new therapies to inhibit and prevent bone metastases. The ideal animal model should be clinically relevant, reproducible, and representative of human disease. Currently, an ideal model does not exist; however, understanding the strengths and weaknesses of the available models will lead to proper study design and successful cancer research. This review provides an overview of the current in vivo animal models used in the study of skeletal metastases or local tumor invasion into bone and focuses on mammary and prostate cancer, lymphoma, multiple myeloma, head and neck squamous cell carcinoma, and miscellaneous tumors that metastasize to bone.
Collapse
Affiliation(s)
- J K Simmons
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - B E Hildreth
- Department of Veterinary Clinical Sciences, The Ohio State University, Columbus, OH, USA
| | - W Supsavhad
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - S M Elshafae
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - B B Hassan
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - W P Dirksen
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| | - R E Toribio
- Department of Veterinary Clinical Sciences, The Ohio State University, Columbus, OH, USA
| | - T J Rosol
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
16
|
Riminucci M, Remoli C, Robey PG, Bianco P. Stem cells and bone diseases: new tools, new perspective. Bone 2015; 70:55-61. [PMID: 25240458 PMCID: PMC5524373 DOI: 10.1016/j.bone.2014.09.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Revised: 09/03/2014] [Accepted: 09/09/2014] [Indexed: 12/13/2022]
Abstract
Postnatal skeletal stem cells are a unique class of progenitors with biological properties that extend well beyond the limits of stemness as commonly defined. Skeletal stem cells sustain skeletal tissue homeostasis, organize and maintain the complex architectural structure of the bone marrow microenvironment and provide a niche for hematopoietic progenitor cells. The identification of stem cells in the human post-natal skeleton has profoundly changed our approach to the physiology and pathology of this system. Skeletal diseases have been long interpreted essentially in terms of defective function of differentiated cells and/or abnormal turnover of the matrix that they produce. The notion of a skeletal stem cell has brought forth multiple, novel concepts in skeletal biology that provide potential alternative concepts. At the same time, the recognition of the complex functions played by skeletal progenitors, such as the structural and functional organization of the bone marrow, has provided an innovative, unifying perspective for understanding bone and bone marrow changes simultaneously occurring in many disorders. Finally, the possibility to isolate and highly enrich for skeletal progenitors, enables us to reproduce perfectly normal or pathological organ miniatures. These, in turn, provide suitable models to investigate and manipulate the pathogenetic mechanisms of many genetic and non-genetic skeletal diseases. This article is part of a Special Issue entitled Stem cells and Bone.
Collapse
Affiliation(s)
- Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, Italy.
| | - Cristina Remoli
- Department of Molecular Medicine, Sapienza University of Rome, Italy
| | - Pamela G Robey
- Craniofacial and Skeletal Diseases Branch, National Institute of Craniofacial and Dental Research, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | - Paolo Bianco
- Department of Molecular Medicine, Sapienza University of Rome, Italy
| |
Collapse
|
17
|
Ye ZB, Ma G, Zhao YH, Xiao Y, Zhan Y, Jing C, Gao K, Liu ZH, Yu SJ. miR-429 inhibits migration and invasion of breast cancer cells in vitro. Int J Oncol 2014; 46:531-8. [PMID: 25405387 PMCID: PMC4277243 DOI: 10.3892/ijo.2014.2759] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 10/23/2014] [Indexed: 12/25/2022] Open
Abstract
Accumulating evidence indicates that microRNAs (miRNAs) are involved in regulating cancer invasion and metastasis, and an increasing number of research demonstrates that miRNAs can promote or inhibit cell motility depending on genetic background of different cancers and the microenvironment. In the present study, we established an in vivo bone metastasis model of breast cancer by injecting MDA-MB-231 cells into the left ventricle of nude mice, and then screened the differentially expressed miRNAs between parental and bone-metastatic MDA-MB-231 cells using miRNA array. The results revealed that decreased expression of miR-429 was probably involved in negatively regulating bone metastasis of breast cancer cells. On the other hand, overexpression of miR-429 in MDA-MB-231 cells remarkably suppressed invasion in vitro. We identified ZEB1 and CRKL as potential targets of miR-429 by analyzing combined results from in silico search and global expression array of the same RNA samples. Immunoblot assay confirmed that miR-429 reduced their expression at protein level. Taken together, our results offer an opportunity for further understanding of the recondite mechanisms underlying the bone metastasis of breast cancer.
Collapse
Affiliation(s)
- Zhi-Bin Ye
- Department of Orthopedics, Cancer Hospital (Institute), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Gang Ma
- The State Key Laboratory of Molecular Oncology, Cancer Hospital (Institute), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Ya-Hui Zhao
- The State Key Laboratory of Molecular Oncology, Cancer Hospital (Institute), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Yun Xiao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150081, P.R. China
| | - Yun Zhan
- The State Key Laboratory of Molecular Oncology, Cancer Hospital (Institute), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Chao Jing
- The State Key Laboratory of Molecular Oncology, Cancer Hospital (Institute), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Kai Gao
- Institute of Laboratory Animal Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Zhi-Hua Liu
- The State Key Laboratory of Molecular Oncology, Cancer Hospital (Institute), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| | - Sheng-Ji Yu
- Department of Orthopedics, Cancer Hospital (Institute), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, P.R. China
| |
Collapse
|
18
|
Simmons JK, Dirksen WP, Hildreth BE, Dorr C, Williams C, Thomas R, Breen M, Toribio RE, Rosol TJ. Canine prostate cancer cell line (Probasco) produces osteoblastic metastases in vivo. Prostate 2014; 74:1251-65. [PMID: 25043424 PMCID: PMC4216720 DOI: 10.1002/pros.22838] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 05/28/2014] [Indexed: 01/07/2023]
Abstract
BACKGROUND In 2012, over 240,000 men were diagnosed with prostate cancer and over 28,000 died from the disease. Animal models of prostate cancer are vital to understanding its pathogenesis and developing therapeutics. Canine models in particular are useful due to their similarities to late-stage, castration-resistant human disease with osteoblastic bone metastases. This study established and characterized a novel canine prostate cancer cell line that will contribute to the understanding of prostate cancer pathogenesis. METHODS A novel cell line (Probasco) was derived from a mixed breed dog that had spontaneous prostate cancer. Cell proliferation and motility were analyzed in vitro. Tumor growth in vivo was studied by subcutaneous, intratibial, and intracardiac injection of Probasco cells into nude mice. Tumors were evaluated by bioluminescent imaging, Faxitron radiography, µCT, and histology. RT-PCR and genome-wide DNA copy number profiling were used to characterize the cell line. RESULTS The Probasco cells grew in vitro (over 75 passages) and were tumorigenic in nude mice. Probasco cells expressed high levels of BMP2, CDH1, MYOF, FOLH1, RUNX2, and SMAD5 modest CXCL12, SLUG, and BMP, and no PTHrP mRNA. Following intracardiac injection, Probasco cells metastasized primarily to the appendicular skeleton, and both intratibial and intracardiac injections produced osteoblastic tumors in bone. Comparative genomic hybridization demonstrated numerous DNA copy number aberrations throughout the genome, including large losses and gains in multiple chromosomes. CONCLUSIONS The Probasco prostate cancer cell line will be a valuable model to investigate the mechanisms of prostate cancer pathogenesis and osteoblastic bone metastases.
Collapse
Affiliation(s)
- Jessica K. Simmons
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio
| | - Wessel P. Dirksen
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio
| | - Blake E. Hildreth
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio
| | - Carlee Dorr
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina
| | - Christina Williams
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina
| | - Rachael Thomas
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina
- Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina
| | - Matthew Breen
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina
- Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina
- Cancer Genetics, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | - Ramiro E. Toribio
- Department of Veterinary Clinical Sciences, The Ohio State University, Columbus, Ohio
| | - Thomas J. Rosol
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio
- Correspondence to: Dr. Thomas J. Rosol, Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio 43210.
| |
Collapse
|
19
|
Orlova VV, van den Hil FE, Petrus-Reurer S, Drabsch Y, Ten Dijke P, Mummery CL. Generation, expansion and functional analysis of endothelial cells and pericytes derived from human pluripotent stem cells. Nat Protoc 2014; 9:1514-31. [PMID: 24874816 DOI: 10.1038/nprot.2014.102] [Citation(s) in RCA: 255] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human endothelial cells (ECs) and pericytes are of great interest for research on vascular development and disease, as well as for future therapy. This protocol describes the efficient generation of ECs and pericytes from human pluripotent stem cells (hPSCs) under defined conditions. Essential steps for hPSC culture, differentiation, isolation and functional characterization of ECs and pericytes are described. Substantial numbers of both cell types can be derived in only 2-3 weeks: this involves differentiation (10 d), isolation (1 d) and 4 or 10 d of expansion of ECs and pericytes, respectively. We also describe two assays for functional evaluation of hPSC-derived ECs: (i) primary vascular plexus formation upon coculture with hPSC-derived pericytes and (ii) incorporation in the vasculature of zebrafish xenografts in vivo. These assays can be used to test the quality and drug sensitivity of hPSC-derived ECs and model vascular diseases with patient-derived hPSCs.
Collapse
Affiliation(s)
- Valeria V Orlova
- 1] Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands. [2] Department of Molecular Cell Biology, Cancer Genomics Centre, Leiden University Medical Center, Leiden, The Netherlands
| | - Francijna E van den Hil
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Sandra Petrus-Reurer
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Yvette Drabsch
- Department of Molecular Cell Biology, Cancer Genomics Centre, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Ten Dijke
- Department of Molecular Cell Biology, Cancer Genomics Centre, Leiden University Medical Center, Leiden, The Netherlands
| | - Christine L Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
20
|
|
21
|
Deng X, He G, Liu J, Luo F, Peng X, Tang S, Gao Z, Lin Q, Keller JM, Yang T, Keller ET. Recent advances in bone-targeted therapies of metastatic prostate cancer. Cancer Treat Rev 2014; 40:730-8. [PMID: 24767837 DOI: 10.1016/j.ctrv.2014.04.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 04/02/2014] [Accepted: 04/07/2014] [Indexed: 10/25/2022]
Abstract
Prostate cancer is one of the most common malignancies affecting men worldwide, with bone being the most common site of metastasis in patients that progress beyond organ confinement. Bone metastases are virtually incurable and result in significant disease morbidity and mortality. Bone provides a unique microenvironment whose local interactions with tumor cells offer novel targets for therapeutic interventions. Several attractive molecules or pathways have been identified as new potential therapeutic targets for bone metastases caused by metastatic castration-resistant prostate cancer. In this review, we present the recent advances in molecular targeted therapies for prostate cancer bone metastasis focusing on therapies that target the bone cells and the bone microenvironment. The therapies covered in this review include agents that inhibit bone resorption, agents that stimulate bone formation, and agents that target the bone matrix. Suggestions to devise more effective molecular targeted therapies are proposed. Hopefully, with better understanding of the biology of the disease and the development of more robust targeted therapies, the survival and quality of life of the affected individuals could be significantly improved.
Collapse
Affiliation(s)
- Xiyun Deng
- College of Medicine, Hunan Normal University, Changsha, Hunan 410013, China; Changsha Microworld Biotech Company, Changsha, Hunan 410004, China
| | - Guangchun He
- College of Medicine, Hunan Normal University, Changsha, Hunan 410013, China
| | - Junwen Liu
- National Engineering Laboratory for Rice and Byproduct In-Depth Processing, Central South University of Forestry and Technology, Changsha, Hunan 410004, China
| | - Feijun Luo
- National Engineering Laboratory for Rice and Byproduct In-Depth Processing, Central South University of Forestry and Technology, Changsha, Hunan 410004, China
| | - Xiaoning Peng
- College of Medicine, Hunan Normal University, Changsha, Hunan 410013, China
| | - Shigang Tang
- College of Medicine, Hunan Normal University, Changsha, Hunan 410013, China
| | - Zhiyong Gao
- College of Medicine, Hunan Normal University, Changsha, Hunan 410013, China
| | - Qinlu Lin
- National Engineering Laboratory for Rice and Byproduct In-Depth Processing, Central South University of Forestry and Technology, Changsha, Hunan 410004, China
| | - Jill M Keller
- Department of Urology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Tao Yang
- National Engineering Laboratory for Rice and Byproduct In-Depth Processing, Central South University of Forestry and Technology, Changsha, Hunan 410004, China; Changsha Microworld Biotech Company, Changsha, Hunan 410004, China.
| | - Evan T Keller
- Department of Urology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
22
|
Colony-stimulating factor 1 potentiates lung cancer bone metastasis. J Transl Med 2014; 94:371-81. [PMID: 24468794 DOI: 10.1038/labinvest.2014.1] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 11/22/2013] [Accepted: 12/22/2013] [Indexed: 11/09/2022] Open
Abstract
Colony-stimulating factor 1 (CSF1) is essential for osteoclastogenesis that mediates osteolysis in metastatic tumors. Patients with lung cancer have increased CSF1 in serum and high levels are associated with poor survival. Adenocarcinomas metastasize rapidly and many patients suffer from bone metastasis. Lung cancer stem-like cells sustain tumor growth and potentiate metastasis. The purpose of this study was to determine the role of CSF1 in lung cancer bone metastasis and whether inhibition of CSF1 ameliorates the disease. Human lung adenocarcinoma A549 cells were examined in vitro for CSF1/CSF1R. A549-luc cells were injected intracardiac in NOD/SCID mice and metastasis was assessed. To determine the effect of CSF1 knockdown (KD) in A549 cells on bone metastasis, cells were stably transfected with a retroviral vector containing short-hairpin CSF1 (KD) or empty vector (CT). Results showed that A549 cells express CSF1/CSF1R; CSF1 increased their proliferation and invasion, whereas soluble CSF1R inhibited invasion. Mice injected with A549-luc cells showed osteolytic bone lesions 3.5 weeks after injection and lesions increased over 5 weeks. Tumors recapitulated adenocarcinoma morphology and showed osteoclasts along the tumor/bone interface, trabecular, and cortical bone loss. Analyses of KD cells showed decreased CSF1 protein levels, reduced colony formation in soft agar assay, and decreased fraction of stem-like cells. In CSF1KD mice, the incidence of tumor metastasis was similar to controls, although fewer CSF1KD mice had metastasis in both hind limbs. KD tumors showed reduced CSF1 expression, Ki-67+ cells, and osteoclasts. Importantly, there was a low incidence of large tumors >0.1 mm(2) in CSF1KD mice compared with control mice (10% vs 62.5%). This study established a lung osteolytic bone metastasis model that resembles human disease and suggests that CSF1 is a key determinant of cancer stem cell survival and tumor growth. Results may lead to novel strategies to inhibit CSF1 in lung cancer and improve management of bone metastasis.
Collapse
|
23
|
Drabsch Y, He S, Zhang L, Snaar-Jagalska BE, ten Dijke P. Transforming growth factor-β signalling controls human breast cancer metastasis in a zebrafish xenograft model. Breast Cancer Res 2013; 15:R106. [PMID: 24196484 PMCID: PMC3978640 DOI: 10.1186/bcr3573] [Citation(s) in RCA: 89] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 10/21/2013] [Indexed: 12/16/2022] Open
Abstract
INTRODUCTION The transforming growth factor beta (TGF-β) signalling pathway is known to control human breast cancer invasion and metastasis. We demonstrate that the zebrafish xenograft assay is a robust and dependable animal model for examining the role of pharmacological modulators and genetic perturbation of TGF-β signalling in human breast tumour cells. METHODS We injected cancer cells into the embryonic circulation (duct of cuvier) and examined their invasion and metastasis into the avascular collagenous tail. Various aspects of the TGF-β signalling pathway were blocked by chemical inhibition, small interfering RNA (siRNA), or small hairpin RNA (shRNA). Analysis was conducted using fluorescent microscopy. RESULTS Breast cancer cells with different levels of malignancy, according to in vitro and in vivo mouse studies, demonstrated invasive and metastatic properties within the embryonic zebrafish model that nicely correlated with their differential tumourigenicity in mouse models. Interestingly, MCF10A M2 and M4 cells invaded into the caudal hematopoietic tissue and were visible as a cluster of cells, whereas MDA MB 231 cells invaded into the tail fin and were visible as individual cells. Pharmacological inhibition with TGF-β receptor kinase inhibitors or tumour specific Smad4 knockdown disturbed invasion and metastasis in the zebrafish xenograft model and closely mimicked the results we obtained with these cells in a mouse metastasis model. Inhibition of matrix metallo proteinases, which are induced by TGF-β in breast cancer cells, blocked invasion and metastasis of breast cancer cells. CONCLUSIONS The zebrafish-embryonic breast cancer xenograft model is applicable for the mechanistic understanding, screening and development of anti-TGF-β drugs for the treatment of metastatic breast cancer in a timely and cost-effective manner.
Collapse
|
24
|
Morisaki T, Umebayashi M, Kiyota A, Koya N, Tanaka H, Onishi H, Katano M. Combining cetuximab with killer lymphocytes synergistically inhibits human cholangiocarcinoma cells in vitro. Anticancer Res 2012; 22:261-71. [PMID: 22641659 DOI: 10.1016/j.semcancer.2012.03.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 03/14/2012] [Accepted: 03/21/2012] [Indexed: 12/31/2022]
Abstract
AIM We explored the possibility of combining adoptive immunotherapy with cytokine-activated killer (CAK) cells and the epidermal growth factor receptor monoclonal antibody, cetuximab, as a treatment for cholangiocarcinoma. MATERIALS AND METHODS CAK cells were cultured with a high-dose of interleukin-2 and anti-CD3 monoclonal antibodies. This cell population contained both activated CD16+/CD56+ (NK) cells and CD3+/NKG2D(high+) T-cells. The effect of CAK cells and cetuximab, alone and in combination, on the viability of human cholangiocarcinoma cells was evaluated. RESULTS Culture of CAK cells alone, but not cetuximab alone, exhibited modest cytotoxicity toward cholangiocarcinoma cells. However, combining CAK cells with cetuximab significantly enhanced cytotoxicity. This enhancement was inhibited by the addition of excess human immunoglobulins, suggesting that antibody-dependent cytotoxicity, mediated by activated NK cells in the CAK cell culture was involved in this mechanism. CONCLUSION Cetuximab may be used to enhance CAK cell therapeutic activity in patients with cholangiocarcinoma, by potentiating antibody-dependent cellular cytotoxicity.
Collapse
Affiliation(s)
- Takashi Morisaki
- Fukuoka General Cancer Clinic, 3-1-1 Sumiyoshi, Hakata-ku, Fukuoka 812-0018, Japan.
| | | | | | | | | | | | | |
Collapse
|
25
|
Hojjat SP, Foltz W, Wise-Milestone L, Whyne CM. Multimodal μCT/μMR based semiautomated segmentation of rat vertebrae affected by mixed osteolytic/osteoblastic metastases. Med Phys 2012; 39:2848-53. [PMID: 22559657 DOI: 10.1118/1.3703590] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
PURPOSE Multimodal microimaging in preclinical models is used to examine the effect of spinal metastases on bony structure; however, the evaluation of tumor burden and its effect on microstructure has thus far been mainly qualitative or semiquantitative. Quantitative analysis of multimodality imaging is a time consuming task, motivating automated methods. As such, this study aimed to develop a low complexity semiautomated multimodal μCT/μMR based approach to segment rat vertebral structure affected by mixed osteolytic/osteoblastic destruction. METHODS Mixed vertebral metastases were developed via intracardiac injection of Ace-1 canine prostate cancer cells in three 4-week-old rnu/rnu rats. μCT imaging (for high resolution bone visualization), T1-weighted μMR imaging (for bone registration), and T2-weighted μMR imaging (for osteolytic tumor visualization) were conducted on one L1, three L2, and one L3 vertebrae (excised). One sample (L1-L3) was processed for undecalcified histology and stained with Goldner's trichome. The μCT and μMR images were registered using a 3D rigid registration algorithm with a mutual information metric. The vertebral microarchitecture was segmented from the μCT images using atlas-based demons deformable registration, levelset curvature evolution, and intensity-based thresholding techniques. The μCT based segmentation contours of the whole vertebrae were used to mask the T2-weighted μMR images, from which the osteolytic tumor tissue was segmented (intensity-based thresholding). RESULTS Accurate registration of μCT and μMRI modalities yielded precise segmentation of whole vertebrae, trabecular centrums, individual trabeculae, and osteolytic tumor tissue. While the algorithm identified the osteoblastic tumor attached to the vertebral pereosteal surfaces, it was limited in segmenting osteoblastic tissue located within the trabecular centrums. CONCLUSIONS This semiautomated segmentation method yielded accurate registration of μCT and μMRI modalities with application to the development of mathematical models analyzing the mechanical stability of metastatically involved vertebrae and in preclinical applications evaluating new and existing treatment effects on tumor burden and skeletal microstructure.
Collapse
Affiliation(s)
- Seyed-Parsa Hojjat
- Orthopedic Biomechanics Laboratory, Sunnybrook Research Institute, Ontario M4N 3M5, Canada.
| | | | | | | |
Collapse
|
26
|
Wise-Milestone L, Akens MK, Rosol TJ, Hojjat SP, Grynpas MD, Whyne CM. Evaluating the effects of mixed osteolytic/osteoblastic metastasis on vertebral bone quality in a new rat model. J Orthop Res 2012; 30:817-23. [PMID: 22025272 DOI: 10.1002/jor.21577] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 10/06/2011] [Indexed: 02/04/2023]
Abstract
Spinal metastases often show mixed areas of enhanced (osteoblastic) bone growth adjacent to areas of thinning (osteolytic) bone. This study aims to quantitatively characterize bone quality and tumor burden within a new rat model of mixed osteolytic/osteoblastic spinal metastases. Mixed vertebral metastases were analyzed in nude rats 21-days post intracardiac injection of Ace-1 canine prostate cancer cells. Vertebral micro-architecture was assessed in µCT images. Histologic processing quantified tumor burden (PTHrP), osteoclast activity (TRAP), and osteoid formation (Goldner's Trichrome) in ½ of all samples. Remaining samples were mechanically tested to failure in compression. Metastatically involved vertebrae exhibited extreme osteolysis, evident through an increase in osteoclasts leading to significantly reduced trabecular bone volume. Metastatically involved vertebrae also exhibited increased osteoid characteristic of osteoblastic lesions. While mechanical properties in tumor-bearing vertebrae were not significantly decreased compared to controls, a strong correlation was found between trabecular volumetric BMD and ultimate force. The highly aggressive Ace-1 skeletal metastases demonstrated predominant osteolysis with some areas of immature, new osteoblastic bone formation. Bone quality resulting from these lesions consisted of decreased structural properties, but without a significant impact on mechanical integrity.
Collapse
Affiliation(s)
- Lisa Wise-Milestone
- Orthopaedic Biomechanics Laboratory, Sunnybrook Research Institute, UB-19, 2075 Bayview Avenue, Toronto, Ontario, Canada M4N 3M5
| | | | | | | | | | | |
Collapse
|
27
|
van der Horst G, van der Pluijm G. Preclinical imaging of the cellular and molecular events in the multistep process of bone metastasis. Future Oncol 2012; 8:415-30. [DOI: 10.2217/fon.12.33] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Bone metastasis is a complex process that ultimately leads to devastating metastatic bone disease. It is therefore of key interest to unravel the mechanisms underlying the multistep process of skeletal metastasis and cancer-induced bone disease, and to develop better treatment and management of patients with this devastating disease. Fortunately, novel technologies are rapidly emerging that allow real-time imaging of molecules, pathogenic processes, drug delivery and drug response in preclinical in vivo models. The outcome of these experimental studies will facilitate clinical cancer research by improving the detection of cancer cell invasion, metastasis and therapy response.
Collapse
Affiliation(s)
- Geertje van der Horst
- Department of Urology, Leiden University Medical Center, J3–100, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Gabri van der Pluijm
- Department of Urology, Leiden University Medical Center, J3–100, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
28
|
van der Horst G, van der Pluijm G. Preclinical models that illuminate the bone metastasis cascade. Recent Results Cancer Res 2012; 192:1-31. [PMID: 22307368 DOI: 10.1007/978-3-642-21892-7_1] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In this chapter currently available preclinical models of tumor progression and bone metastasis, including genetically engineered mice that develop primary and metastatic carcinomas and transplantable animal models, will be described. Understanding the multistep process of incurable bone metastasis is pivotal to the development of new therapeutic strategies. Novel technologies for imaging molecules or pathologic processes in cancers and their surrounding stroma have emerged rapidly and have greatly facilitated cancer research, in particular the cellular behavior of osteotropic tumors and their response to new and existing therapeutic agents. Optical imaging, in particular, has become an important tool in preclinical bone metastasis models, clinical trials and medical practice. Advances in experimental and clinical imaging will-in the long run-result in significant improvements in diagnosis, tumor localization, enhanced drug delivery and treatment.
Collapse
|
29
|
Identification and functional validation of therapeutic targets for malignant melanoma. Crit Rev Oncol Hematol 2009; 72:194-214. [DOI: 10.1016/j.critrevonc.2009.02.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2008] [Revised: 02/04/2009] [Accepted: 02/19/2009] [Indexed: 12/12/2022] Open
|
30
|
|
31
|
Lai CL, van den Ham R, Mol J, Teske E. Immunostaining of the androgen receptor and sequence analysis of its DNA-binding domain in canine prostate cancer. Vet J 2008; 181:256-60. [PMID: 18583166 DOI: 10.1016/j.tvjl.2008.04.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2008] [Revised: 03/29/2008] [Accepted: 04/13/2008] [Indexed: 11/29/2022]
Abstract
Prostate cancer in the dog (cPC) has many features in common with hormone refractory human prostate cancer. As cPC is seen more often in castrated dogs, the contribution of the androgen receptor (AR) to the development of prostate cancer remains questionable. The aim of the present study was to evaluate the presence of the AR by immunohistochemistry in cPC. AR staining was observed in most tumors from intact and castrated dogs, but the proportion of positive cells and the staining intensity were much lower than in the prostate of healthy, non-castrated dogs. Most of the positive staining was seen in the cytoplasm rather than in the nuclei of the tumor cells. The predominant cytoplasmic localization was not related to mutations in exon 3 of the DNA-binding domain of the AR, as shown by sequence analysis of microdissected AR positive tumor cells. Other mechanisms that lead to an impaired androgen-AR signaling or a basal/stem cell like origin may explain the low cytoplasmic AR staining in cPC.
Collapse
Affiliation(s)
- Chen-Li Lai
- Department of Clinical Sciences of Companion Animals, Faculty of Veterinary Medicine, Utrecht University, P.O. Box 80.154, 3508 TD Utrecht, The Netherlands
| | | | | | | |
Collapse
|
32
|
Henriquez NV, van Overveld PGM, Que I, Buijs JT, Bachelier R, Kaijzel EL, Löwik CWGM, Clezardin P, van der Pluijm G. Advances in optical imaging and novel model systems for cancer metastasis research. Clin Exp Metastasis 2007; 24:699-705. [PMID: 17972147 DOI: 10.1007/s10585-007-9115-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Accepted: 10/10/2007] [Indexed: 10/22/2022]
Abstract
Research into the genetic and physiological interactions of tumours with their host environment requires in vivo assays to address molecular expression patterns and function. In recent years much of this work has been performed using bioluminescent and fluorescent imaging techniques that allow real-time and non-invasive imaging of gene expression and (tumour) tissue development. Luminescence imaging has until now been more or less the only tool that allows the imaging of intra-osseous breast cancer cells and indeed this technique has been pioneered in our laboratory. Here we summarise some recent innovations and developments using cancer cells and some of the first imaging models of multimodal dual luminescence and luminescence combined with fluorescence of intra-osseous tumours. We further engineered our models to incorporate a specific insertion site in the genome and will discuss some of the possible applications. These include the insertion of signalling pathway-specific reporters and studying the fate of multiple injected populations in a single mouse. We conclude that recent improvements in luminescence- and fluorescence-detection platforms now clearly allow multimodal imaging which will greatly enhance our ability to assess gene function and for the first time to visualise multiple gene- and cellular interactions in real time and in vivo.
Collapse
Affiliation(s)
- Nico V Henriquez
- Department of Endocrinology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Kaijzel EL, van der Pluijm G, Löwik CWGM. Whole-body optical imaging in animal models to assess cancer development and progression. Clin Cancer Res 2007; 13:3490-7. [PMID: 17575211 DOI: 10.1158/1078-0432.ccr-07-0402] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Different optical-based imaging models were used to investigate tumor progression and metastasis with particular emphasis on metastasis to bone and bone marrow. We describe how optical imaging can be used to follow important processes in tumor development and treatment response, including angiogenesis, apoptosis, and proteolysis. Finally, we discuss the translation of one optical imaging modality, near-IR fluorescence, from animal validation studies to applications in the clinic related to cancer management.
Collapse
Affiliation(s)
- Eric L Kaijzel
- Departments of Endocrinology and Metabolic Diseases and Urology, Leiden University Medical Center, Leiden, the Netherlands
| | | | | |
Collapse
|
34
|
Vernon AE, Bakewell SJ, Chodosh LA. Deciphering the molecular basis of breast cancer metastasis with mouse models. Rev Endocr Metab Disord 2007; 8:199-213. [PMID: 17657606 DOI: 10.1007/s11154-007-9041-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Breast cancer begins as a localized disease, but has the potential to spread to distant sites within the body. This process--known as metastasis--is the leading cause of death from breast cancer. Whether the ability of cancer cells to metastasize is an intrinsic or acquired feature is currently a topic of considerable debate. Nevertheless, the key cellular events required for metastasis are generally accepted. These include invasion of the surrounding stromal tissue, intravasation, evasion of programmed cell death, arrest within the vasculature at a distant site, extravasation, and establishment and growth within a new microenvironment. The development of mouse models that faithfully mimic critical aspects of human neoplasia has been instrumental in framing our current understanding of multistage carcinogenesis. This review examines the advantages and limitations of existing murine models for mammary carcinogenesis for probing the molecular mechanisms that contribute to metastasis, as well as non-invasive tumor imaging approaches to facilitate these investigations.
Collapse
Affiliation(s)
- Ann E Vernon
- Department of Cancer Biology, and The Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, 612 BRB II/III, 421 Curie Boulevard, Philadelphia, PA 19104-6160, USA
| | | | | |
Collapse
|
35
|
Hermiston TW, Kirn DH. Genetically based therapeutics for cancer: similarities and contrasts with traditional drug discovery and development. Mol Ther 2005; 11:496-507. [PMID: 15771953 DOI: 10.1016/j.ymthe.2004.12.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2004] [Accepted: 12/07/2004] [Indexed: 11/22/2022] Open
Abstract
The field of molecular therapeutics is in its infancy and represents a promising and novel avenue for targeted cancer treatments. Like the small-molecule and antibody therapeutics before them, however, the genetic-based therapies will face significant research and development challenges in their maturation toward an approved cancer therapy. To facilitate this process, we outline and examine in this review the drug development process, briefly summarizing the research and development paradigms that have accompanied the recent successes of the small-molecule and antibody-based cancer therapeutics. Using this background, we compare and contrast the research and development experiences of small-molecule and antibody therapeutics with genetic-based cancer therapeutics, using oncolytic viruses as a defined example of an experimental molecular therapeutic for cancer.
Collapse
Affiliation(s)
- Terry W Hermiston
- Department of Gene Therapy, Berlex Biosciences, Richmond, CA 94941, USA.
| | | |
Collapse
|
36
|
Schneider A, Kalikin LM, Mattos AC, Keller ET, Allen MJ, Pienta KJ, McCauley LK. Bone turnover mediates preferential localization of prostate cancer in the skeleton. Endocrinology 2005; 146:1727-36. [PMID: 15637291 DOI: 10.1210/en.2004-1211] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Bone metastasis is a common untreatable complication associated with prostate cancer. Metastatic cells seed in skeletal sites under active turnover containing dense marrow cellularity. We hypothesized that differences in these skeletal-specific processes are among the critical factors that facilitate the preferential localization of metastatic prostate cancer in bone. To test this, athymic mice were administered PTH to induce bone turnover and increase marrow cellularity daily 1 wk before and after intracardiac inoculation of luciferase-tagged PC-3 cells. Tumor localization was monitored by bioluminescence imaging weekly for 5 wk. At the time of tumor inoculation, PTH-treated mice demonstrated significant increases in serum levels of bone turnover markers such as osteocalcin and tartrate-resistant acid phosphatase 5b and in the number of tartrate-resistant acid phosphatase-positive osteoclasts per millimeter of bone when compared with the other groups. Likewise, PTH treatment stimulated a qualitative increase in marrow cellular proliferation as determined by 5-bromo-2'-deoxyuridine immunostaining. Skeletal metastases formed in the hind limb and craniofacial regions of young mice with no difference between groups. In adult mice, however, bioluminescent signals in the hind limb and craniofacial regions were 3-fold higher in PTH-treated mice vs. controls. Fluorochrome labeling revealed increased bone formation activity in trabecular bone adjacent to tumors. When zoledronic acid, a nitrogen-containing bisphosphonate that inhibits osteoclast-mediated bone resorption, was administered concurrently with PTH, a significant reduction in the incidence of bone tumors was observed. Overall, these studies provide new evidence that skeletal sites rich in marrow cellularity under active turnover offer a more congenial microenvironment to facilitate cancer localization in the skeleton.
Collapse
Affiliation(s)
- Abraham Schneider
- Department of Periodontics/Prevention/Geriatrics, School of Dentistry, Room 3343, University of Michigan, 1011 North University Avenue, Ann Arbor, Michigan 48109-1078, USA
| | | | | | | | | | | | | |
Collapse
|