1
|
Morita M, Arshad F, Quayle LA, George CN, Lefley DV, Kalajzic I, Balsubramanian M, Cebe T, Reilly G, Bishop NJ, Ottewell PD. Losartan alters osteoblast differentiation and increases bone mass through inhibition of TGF B signalling in vitro and in an OIM mouse model. Bone Rep 2024; 22:101795. [PMID: 39185375 PMCID: PMC11344016 DOI: 10.1016/j.bonr.2024.101795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/25/2024] [Accepted: 07/22/2024] [Indexed: 08/27/2024] Open
Abstract
Excessive production of Transforming Growth Factor β (TGFβ) is commonly associated with dominant and recessive forms of OI. Previous reports have indicated that administration of TGFβ-targeted antibodies maybe of potential therapeutic benefit to OI patients. However, direct targeting of TGFβ is likely to cause multiple adverse effects including simulation of autoimmunity. In the current study we use patient-derived normal and OI fibroblasts, osteoblasts and OIM mouse models to determine the effects of Losartan, an angiotensin II receptor type 1 (AT1) antagonist, on TGFβ signalling and bone morphology in OI. In OIM mice bred on a mixed background administration of 0.6 g/L losartan for 4 weeks was associated with a significant reduction in TGFβ from 79.2 g/L in the control to 60.0 ng/ml following losartan (p < 0.05), reduced osteoclast activity as measured by CTX from 275.9 ng/ml in the control to 157.2 ng/ml following 0.6 g/L of losartan (p < 0.05) and increased cortical bone thickness (P < 0.001). Furthermore in OIM mice bred on a C57BL/6 background 0.6 g/L losartan increased trabecular bone volume in the tibiae (P < 0.05) and the vertebrae (P < 0.01), increased cortical bone thickness (P < 0.001) reduced the trabecular pattern factor (P < 0.01 and P < 0.001 for the tibiae and vertebrae respectively), reduced osteoclast (P < 0.05) and osteoblast (P < 0.01) numbers as well as reducing the area of bone covered by these cell types. Interestingly, losartan did not affect immune cells infiltrating into bone, nor did this drug alter TGFβ signalling in normal or OI fibroblasts. Instead, losartan reduced SMAD2 phosphorylation in osteoblasts, inhibiting their ability to differentiate. Our data suggest that losartan may be an effective treatment for the bone-associated dysmorphia displayed in OI whilst minimising potential adverse immune cell-related effects.
Collapse
Affiliation(s)
- Mai Morita
- Mellanby Centre for Musculoskeletal Research and Division of Clinical Medicine, University of Sheffield, Beech Hill Road, Sheffield, UK
| | - Fawaz Arshad
- Mellanby Centre for Musculoskeletal Research and Division of Clinical Medicine, University of Sheffield, Beech Hill Road, Sheffield, UK
| | - Lewis A. Quayle
- Department of Computing, Sheffield Hallam University, Cantor Building, Arundel Street, Sheffield, UK
| | - Christopher N. George
- Mellanby Centre for Musculoskeletal Research and Division of Clinical Medicine, University of Sheffield, Beech Hill Road, Sheffield, UK
| | - Diane V. Lefley
- Mellanby Centre for Musculoskeletal Research and Division of Clinical Medicine, University of Sheffield, Beech Hill Road, Sheffield, UK
| | - Ivo Kalajzic
- Reconstructive Sciences, UConn Health, Farmington, CT. USA
| | - Meena Balsubramanian
- Mellanby Centre for Musculoskeletal Research and Division of Clinical Medicine, University of Sheffield, Beech Hill Road, Sheffield, UK
- Highly specialised Osteogenesis Imparfecta Service, Sheffield Children's NHS Foundation Trust, Sheffield, UK
| | - Tugba Cebe
- INSIGNIO Institute for in silico Medicine and the Kroto Research Institute, Department of Materials Science and Engineering, University of Sheffield, Sheffield, UK
| | - Gwen Reilly
- INSIGNIO Institute for in silico Medicine and the Kroto Research Institute, Department of Materials Science and Engineering, University of Sheffield, Sheffield, UK
| | - Nicolas J. Bishop
- Mellanby Centre for Musculoskeletal Research and Division of Clinical Medicine, University of Sheffield, Beech Hill Road, Sheffield, UK
- Highly specialised Osteogenesis Imparfecta Service, Sheffield Children's NHS Foundation Trust, Sheffield, UK
| | - Penelope D. Ottewell
- Mellanby Centre for Musculoskeletal Research and Division of Clinical Medicine, University of Sheffield, Beech Hill Road, Sheffield, UK
| |
Collapse
|
2
|
Novel Methods of Targeting IL-1 Signalling for the Treatment of Breast Cancer Bone Metastasis. Cancers (Basel) 2022; 14:cancers14194816. [PMID: 36230739 PMCID: PMC9561984 DOI: 10.3390/cancers14194816] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The pro-inflammatory cytokine, IL1β, plays a pivotal role in breast cancer bone metastasis. Inhibiting IL-1 signalling with the IL1β specific antibody, Canakinumab, or the IL1R1 antagonist Anakinra almost eliminates bone metastases but has adverse effects on tumours growing outside of the bone and immune regulation. This current study demonstrated that pharmacological inhibition of other members of the IL-1 signalling pathway Caspase-1, IL1β and IL1R reduced migration and invasion of E0771 and Py8119 cells in vitro and also reduced spontaneous metastasis and metastatic outgrowth of breast cancer in the bone, in vivo. Interestingly, targeting IRAK1 had no anti-tumour effects. Importantly, inhibiting Caspase-1 reduces bone metastasis without adversely affecting tumours outside of bone or immune cell regulation, suggesting that targeting immediately upstream of IL1β may be a good therapeutic strategy for treating patients with breast-cancer-induced bone disease. Abstract Breast cancer bone metastasis is currently incurable. Evidence suggests that inhibiting IL-1 signalling with the IL1R antagonist, Anakinra, or the IL1β antibody, Canakinumab, prevents metastasis and almost eliminates breast cancer growth in the bone. However, these drugs increase primary tumour growth. We, therefore, investigated whether targeting other members of the IL-1 pathway (Caspase-1, IL1β or IRAK1) could reduce bone metastases without increasing tumour growth outside of the bone. Inhibition of IL-1 via MLX01 (IL1β secretion inhibitor), VRT043198/VX765 (Caspase-1 inhibitor), Pacritinib (IRAK1 inhibitor) or Anakinra (IL1R antagonist) on tumour cell viability, migration and invasion were assessed in mouse mammary E0771 and Py8119 cells in vitro and on primary tumour growth, spontaneous metastasis and metastatic outgrowth in vivo. In vitro, Inhibition of IL-1 signalling by MLX01, VRT043198 and Anakinra reduced migration of E0771 and Py8119 cells and reversed tumour-derived IL1β induced-increased invasion and migration towards bone cells. In vivo, VX765 and Anakinra significantly reduced spontaneous metastasis and metastatic outgrowth in the bone, whereas MLX01 reduced primary tumour growth and bone metastasis. Pacritinib had no effect on metastasis in vitro or in vivo. Targeting IL-1 signalling with small molecule inhibitors may provide a new therapeutic strategy for breast cancer bone metastasis.
Collapse
|
3
|
Tulotta C, Lefley DV, Moore CK, Amariutei AE, Spicer-Hadlington AR, Quayle LA, Hughes RO, Ahmed K, Cookson V, Evans CA, Vadakekolathu J, Heath P, Francis S, Pinteaux E, Pockley AG, Ottewell PD. IL-1B drives opposing responses in primary tumours and bone metastases; harnessing combination therapies to improve outcome in breast cancer. NPJ Breast Cancer 2021; 7:95. [PMID: 34290237 PMCID: PMC8295314 DOI: 10.1038/s41523-021-00305-w] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 07/01/2021] [Indexed: 12/20/2022] Open
Abstract
Breast cancer bone metastasis is currently incurable, ~75% of patients with late-stage breast cancer develop disease recurrence in bone and available treatments are only palliative. We have previously shown that production of the pro-inflammatory cytokine interleukin-1B (IL-1B) by breast cancer cells drives bone metastasis in patients and in preclinical in vivo models. In the current study, we have investigated how IL-1B from tumour cells and the microenvironment interact to affect primary tumour growth and bone metastasis through regulation of the immune system, and whether targeting IL-1 driven changes to the immune response improves standard of care therapy for breast cancer bone metastasis. Using syngeneic IL-1B/IL1R1 knock out mouse models in combination with genetic manipulation of tumour cells to overexpress IL-1B/IL1R1, we found that IL-1B signalling elicited an opposite response in primary tumours compared with bone metastases. In primary tumours, IL-1B inhibited growth, by impairing the infiltration of innate immune cell subsets with potential anti-cancer functions but promoted enhanced tumour cell migration. In bone, IL-1B stimulated the development of osteolytic metastases. In syngeneic models of breast cancer, combining standard of care treatments (Doxorubicin and Zoledronic acid) with the IL-1 receptor antagonist Anakinra inhibited both primary tumour growth and metastasis. Anakinra had opposite effects on the immune response compared to standard of care treatment, and its anti-inflammatory signature was maintained in the combination therapy. These data suggest that targeting IL-1B signalling may provide a useful therapeutic approach to inhibit bone metastasis and improve efficacy of current treatments for breast cancer patients.
Collapse
Affiliation(s)
- Claudia Tulotta
- Department of Oncology and Metabolism, Weston Park Cancer Centre, University of Sheffield, Sheffield, UK
| | - Diane V Lefley
- Department of Oncology and Metabolism, Weston Park Cancer Centre, University of Sheffield, Sheffield, UK
| | - Charlotte K Moore
- Department of Oncology and Metabolism, Weston Park Cancer Centre, University of Sheffield, Sheffield, UK
| | - Ana E Amariutei
- Department of Oncology and Metabolism, Weston Park Cancer Centre, University of Sheffield, Sheffield, UK
| | - Amy R Spicer-Hadlington
- Department of Oncology and Metabolism, Weston Park Cancer Centre, University of Sheffield, Sheffield, UK
| | - Lewis A Quayle
- Department of Oncology and Metabolism, Weston Park Cancer Centre, University of Sheffield, Sheffield, UK
| | - Russell O Hughes
- Department of Oncology and Metabolism, Weston Park Cancer Centre, University of Sheffield, Sheffield, UK
| | - Khawla Ahmed
- Department of Oncology and Metabolism, Weston Park Cancer Centre, University of Sheffield, Sheffield, UK
| | - Victoria Cookson
- Department of Oncology and Metabolism, Weston Park Cancer Centre, University of Sheffield, Sheffield, UK
| | - Catherine A Evans
- Department of Oncology and Metabolism, Weston Park Cancer Centre, University of Sheffield, Sheffield, UK
| | - Jayakumar Vadakekolathu
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Paul Heath
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, UK
| | - Sheila Francis
- Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, UK
| | - Emmanuel Pinteaux
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - A Graham Pockley
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Penelope D Ottewell
- Department of Oncology and Metabolism, Weston Park Cancer Centre, University of Sheffield, Sheffield, UK.
| |
Collapse
|
4
|
Canuas-Landero VG, George CN, Lefley DV, Corness H, Muthana M, Wilson C, Ottewell PD. Oestradiol Contributes to Differential Antitumour Effects of Adjuvant Zoledronic Acid Observed Between Pre- and Post-Menopausal Women. Front Endocrinol (Lausanne) 2021; 12:749428. [PMID: 34733240 PMCID: PMC8559775 DOI: 10.3389/fendo.2021.749428] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/20/2021] [Indexed: 12/26/2022] Open
Abstract
Clinical trials have demonstrated that adding zoledronic acid (Zol) to (neo)adjuvant standard of care has differential antitumour effects in pre- and post-menopausal women: Both benefit from reduced recurrence in bone; however, while postmenopausal women also incur survival benefit, none is seen in premenopausal women treated with adjuvant bisphosphonates. In the current study, we have used mouse models to investigate the role of oestradiol in modulating potential antitumour effects of Zol. Pre-, peri-, and post-menopausal concentrations of oestradiol were modelled in BALB/c wild-type, BALB/c nude, and C57BL/6 mice by ovariectomy followed by supplementation with oestradiol. Mice also received 40 mg/kg/day goserelin to prevent ovariectomy-induced increases in follicle-stimulating hormone (FSH). Metastasis was modelled following injection of MDA-MB-231, 4T1, or E0771 cells after ovariectomy and saline or 100 μg/kg Zol administered weekly. Supplementing ovariectomised mice with 12.5 mg/ml, 1.38 mg/ml, and 0 ng/ml oestradiol, in the presence of goserelin, resulted in serum concentrations of 153.16 ± 18.10 pg/ml, 48.64 ± 18.44 pg/ml, and 1.00 ± 0.27 pg/ml oestradiol, which are equivalent to concentrations found in pre-, peri-, and post-menopausal humans. Osteoclast activity was increased 1.5-1.8-fold with peri- and post-menopausal compared with premenopausal oestradiol, resulting in a 1.34-1.69-fold reduction in trabecular bone. Zol increased trabecular bone in all groups but did not restore bone to volumes observed under premenopausal conditions. In tumour-bearing mice, Zol reduced bone metastases in BALB/c (wild-type and nude), with greatest effects seen under pre- and post-menopausal concentrations of oestradiol. Zol did not affect soft tissue metastases in immunocompetent BALB/c mice but increased metastases 3.95-fold in C57BL/6 mice under premenopausal concentrations of oestradiol. In contrast, Zol significantly reduced soft tissue metastases 2.07 and 4.69-fold in immunocompetent BALB/c and C57BL/6 mice under postmenopausal oestradiol, mirroring the results of the clinical trials of (neo)adjuvant bisphosphonates. No effects on soft tissue metastases were observed in immunocompromised mice, and differences in antitumour response did not correlate with musculoaponeurotic fibrosarcoma (MAF), macrophage capping protein (CAPG), or PDZ domain containing protein GIPC1 (GIPC1) expression. In conclusion, oestradiol contributes to altered antitumour effects of Zol observed between pre- and post-menopausal women. However, other immunological/microenvironmental factors are also likely to contribute to this phenomenon.
Collapse
|
5
|
Yao Z, Murali B, Ren Q, Luo X, Faget DV, Cole T, Ricci B, Thotala D, Monahan J, van Deursen JM, Baker D, Faccio R, Schwarz JK, Stewart SA. Therapy-Induced Senescence Drives Bone Loss. Cancer Res 2020; 80:1171-1182. [PMID: 31932453 PMCID: PMC7056549 DOI: 10.1158/0008-5472.can-19-2348] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/14/2019] [Accepted: 12/23/2019] [Indexed: 02/01/2023]
Abstract
Chemotherapy is important for cancer treatment, however, toxicities limit its use. While great strides have been made to ameliorate the acute toxicities induced by chemotherapy, long-term comorbidities including bone loss remain a significant problem. Chemotherapy-driven estrogen loss is postulated to drive bone loss, but significant data suggests the existence of an estrogen-independent mechanism of bone loss. Using clinically relevant mouse models, we showed that senescence and its senescence-associated secretory phenotype (SASP) contribute to chemotherapy-induced bone loss that can be rescued by depleting senescent cells. Chemotherapy-induced SASP could be limited by targeting the p38MAPK-MK2 pathway, which resulted in preservation of bone integrity in chemotherapy-treated mice. These results transform our understanding of chemotherapy-induced bone loss by identifying senescent cells as major drivers of bone loss and the p38MAPK-MK2 axis as a putative therapeutic target that can preserve bone and improve a cancer survivor's quality of life. SIGNIFICANCE: Senescence drives chemotherapy-induced bone loss that is rescued by p38MAPK or MK2 inhibitors. These findings may lead to treatments for therapy-induced bone loss, significantly increasing quality of life for cancer survivors.
Collapse
Affiliation(s)
- Zhangting Yao
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Bhavna Murali
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Qihao Ren
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Xianmin Luo
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Douglas V Faget
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Tom Cole
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri
| | - Biancamaria Ricci
- Department of Orthopaedics, Washington University School of Medicine, St. Louis, Missouri
| | - Dinesh Thotala
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri
| | | | - Jan M van Deursen
- Department of Biochemistry and Molecular Biology and Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota
| | - Darren Baker
- Department of Biochemistry and Molecular Biology and Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota
| | - Roberta Faccio
- Department of Orthopaedics, Washington University School of Medicine, St. Louis, Missouri
- Shriners Hospital for Children, St. Louis, Missouri
| | - Julie K Schwarz
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, Missouri
| | - Sheila A Stewart
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri.
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, Missouri
- ICCE Institute, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
6
|
Hughes R, Chen X, Hunter KD, Hobbs JK, Holen I, Brown NJ. Bone marrow osteoprogenitors are depleted whereas osteoblasts are expanded independent of the osteogenic vasculature in response to zoledronic acid. FASEB J 2019; 33:12768-12779. [PMID: 31490705 PMCID: PMC6902700 DOI: 10.1096/fj.201900553rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 08/05/2019] [Indexed: 12/21/2022]
Abstract
Zoledronic acid (ZOL) is an antiresorptive drug used to prevent bone loss in a variety of conditions, acting mainly through suppression of osteoclast activity. There is growing evidence that ZOL can also affect cells of the mesenchymal lineage in bone. We present novel data revealing significant changes in the abundance of perivascular mesenchymal stromal cells (MSCs)/osteoprogenitors and osteoblasts following the injection of ZOL, in vivo. In young mice with high bone turnover and an abundance of perivascular osteoprogenitors, ZOL significantly (P < 0.0001) increased new bone formation. This was accompanied by a decline in osterix-positive osteoprogenitors and a corresponding increase in osteoblasts. However, these effects were not observed in mature mice with low bone turnover. Interestingly, the ZOL-induced changes in cells of the mesenchymal lineage occurred independently of effects on the osteogenic vasculature. Thus, we demonstrate that a single, clinically relevant dose of ZOL can induce new bone formation in microenvironments enriched for perivascular MSC/osteoprogenitors and high osteogenic potential. This arises from the differentiation of perivascular osterix-positive MSC/osteoprogenitors into osteoblasts at sites that are innately osteogenic. Collectively, our data demonstrate that ZOL affects multiple cell types in bone and has differential effects depending on the level of bone turnover.-Hughes, R., Chen, X., Hunter, K. D., Hobbs, J. K., Holen, I., Brown, N. J. Bone marrow osteoprogenitors are depleted whereas osteoblasts are expanded independent of the osteogenic vasculature in response to zoledronic acid.
Collapse
Affiliation(s)
- Russell Hughes
- Department of Oncology and Metabolism, Experimental Cancer Medicine Centre, University of Sheffield, Sheffield, United Kingdom
| | - Xinyue Chen
- Department of Oncology and Metabolism, Experimental Cancer Medicine Centre, University of Sheffield, Sheffield, United Kingdom
- Department of Physics and Astronomy, University of Sheffield, Sheffield, United Kingdom
| | - Keith D. Hunter
- School of Clinical Dentistry, University of Sheffield, United Kingdom
| | - Jamie K. Hobbs
- Department of Physics and Astronomy, University of Sheffield, Sheffield, United Kingdom
| | - Ingunn Holen
- Department of Oncology and Metabolism, Experimental Cancer Medicine Centre, University of Sheffield, Sheffield, United Kingdom
| | - Nicola J. Brown
- Department of Oncology and Metabolism, Experimental Cancer Medicine Centre, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
7
|
Abstract
PURPOSE OF REVIEW The goal of this review is to summarize recent experimental and clinical evidence for metastatic latency and the molecular mechanisms that regulate tumor dormancy in the bone. RECENT FINDINGS Tumor dormancy contributes to the progression of metastasis and thus has significant clinical implications for prognosis and treatment. Tumor-intrinsic signaling and specialized bone marrow niches play a pivotal role in determining the dormancy status of bone disseminated tumor cells. Experimental models have provided significant insight into the effects of the bone microenvironment on tumor cells; however, these models remain limited in their ability to study dormancy. Despite recent advances in the mechanistic understanding of how tumor cells remain dormant in the bone for prolonged periods of time, the signals that trigger spontaneous dormancy escape remain unclear. This review highlights the need for further investigation of mechanisms underlying tumor dormancy using clinically relevant models.
Collapse
Affiliation(s)
- Miranda E Clements
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Rachelle W Johnson
- Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, 37232, USA.
| |
Collapse
|
8
|
Ottewell P, Wilson C. Bone-Targeted Agents in Breast Cancer: Do We Now Have All the Answers? BREAST CANCER-BASIC AND CLINICAL RESEARCH 2019; 13:1178223419843501. [PMID: 31555046 PMCID: PMC6751527 DOI: 10.1177/1178223419843501] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 12/23/2022]
Abstract
The bone-targeted agents (BTAs), bisphosphonates and denosumab, have an established role in the treatment of metastatic breast cancer bone disease and the prevention of cancer-treatment-induced bone loss. Evidence in support of their ability to improve survival in early breast cancer now indicates that the bisphosphonates are effective in postmenopausal women (naturally or chemically induced), but denosumab does not have similar benefits when added to standard adjuvant therapy. In postmenopausal women with early breast cancer, the choice of BTA may differ depending on the indication for treatment; for fracture prevention in low disease recurrence risk patients, denosumab may be favoured (in comparison with placebo) to maintain bone health, and when disease recurrence prevention is a priority in higher risk patients, bisphosphonates may be favoured. The reason for the lack of efficacy of BTAs in premenopausal/perimenopausal patients still remains unanswered and will need preclinical research to evaluate novel treatment combinations with BTAs in this patient group. This review covers the past, present, and future indications for BTAs in both metastatic and early breast cancer.
Collapse
Affiliation(s)
- Penny Ottewell
- Department of Oncology & Metabolism, The Medical School, The University of Sheffield, Sheffield, UK
| | - Caroline Wilson
- Weston Park Cancer Hospital, Sheffield Teaching Hospitals NHS Trust, UK
| |
Collapse
|
9
|
Tulotta C, Lefley DV, Freeman K, Gregory WM, Hanby AM, Heath PR, Nutter F, Wilkinson JM, Spicer-Hadlington AR, Liu X, Bradbury SMJ, Hambley L, Cookson V, Allocca G, Kruithof de Julio M, Coleman RE, Brown JE, Holen I, Ottewell PD. Endogenous Production of IL1B by Breast Cancer Cells Drives Metastasis and Colonization of the Bone Microenvironment. Clin Cancer Res 2019; 25:2769-2782. [PMID: 30670488 DOI: 10.1158/1078-0432.ccr-18-2202] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 11/20/2018] [Accepted: 01/17/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Breast cancer bone metastases are incurable, highlighting the need for new therapeutic targets. After colonizing bone, breast cancer cells remain dormant, until signals from the microenvironment stimulate outgrowth into overt metastases. Here we show that endogenous production of IL1B by tumor cells drives metastasis and growth in bone. EXPERIMENTAL DESIGN Tumor/stromal IL1B and IL1 receptor 1 (IL1R1) expression was assessed in patient samples and effects of the IL1R antagonist, Anakinra, or the IL1B antibody canakinumab on tumor growth and spontaneous metastasis were measured in a humanized mouse model of breast cancer bone metastasis. Effects of tumor cell-derived IL1B on bone colonization and parameters associated with metastasis were measured in MDA-MB-231, MCF7, and T47D cells transfected with IL1B/control. RESULTS In tissue samples from >1,300 patients with stage II/III breast cancer, IL1B in tumor cells correlated with relapse in bone (HR = 1.85; 95% CI, 1.05-3.26; P = 0.02) and other sites (HR = 2.09; 95% CI, 1.26-3.48; P = 0.0016). In a humanized model of spontaneous breast cancer metastasis to bone, Anakinra or canakinumab reduced metastasis and reduced the number of tumor cells shed into the circulation. Production of IL1B by tumor cells promoted epithelial-to-mesenchymal transition (altered E-Cadherin, N-Cadherin, and G-Catenin), invasion, migration, and bone colonization. Contact between tumor and osteoblasts or bone marrow cells increased IL1B secretion from all three cell types. IL1B alone did not stimulate tumor cell proliferation. Instead, IL1B caused expansion of the bone metastatic niche leading to tumor proliferation. CONCLUSIONS Pharmacologic inhibition of IL1B has potential as a novel treatment for breast cancer metastasis.
Collapse
Affiliation(s)
- Claudia Tulotta
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Diane V Lefley
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Katy Freeman
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Walter M Gregory
- Leeds Institute of Clinical Trials Research, Leeds, United Kingdom
| | - Andrew M Hanby
- Institute of Molecular Medicine, St James's University Hospital, Leeds, United Kingdom
| | - Paul R Heath
- Sheffield Institute for Translational Neuroscience, University of Sheffield, Sheffield, United Kingdom
| | - Faith Nutter
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - J Mark Wilkinson
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | | | - Xinming Liu
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Steven M J Bradbury
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Lisa Hambley
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Victoria Cookson
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Gloria Allocca
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | | | - Robert E Coleman
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Janet E Brown
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Ingunn Holen
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Penelope D Ottewell
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
10
|
Tulotta C, Groenewoud A, Snaar-Jagalska BE, Ottewell P. Animal Models of Breast Cancer Bone Metastasis. Methods Mol Biol 2019; 1914:309-330. [PMID: 30729473 DOI: 10.1007/978-1-4939-8997-3_17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
This chapter is designed to provide a comprehensive overview outlining the different in vivo models available for research into breast cancer bone metastasis. The main focus is to guide the researcher through the methodological processes required to establish and utilize these models within their own laboratory. These detailed methods are designed to enable the acquisition of accurate and meaningful results that can be used for publication and future translation into clinical benefit for women with breast cancer-induced bone metastasis.
Collapse
Affiliation(s)
- Claudia Tulotta
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, UK
| | - Arwin Groenewoud
- Institute of Biology, Leiden University, Leiden, The Netherlands
| | | | - Penelope Ottewell
- Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, UK.
| |
Collapse
|
11
|
Holen I, Lefley DV, Francis SE, Rennicks S, Bradbury S, Coleman RE, Ottewell P. IL-1 drives breast cancer growth and bone metastasis in vivo. Oncotarget 2018; 7:75571-75584. [PMID: 27765923 PMCID: PMC5342762 DOI: 10.18632/oncotarget.12289] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 09/15/2016] [Indexed: 01/19/2023] Open
Abstract
Background We have recently identified interleukin 1B (IL-1B) as a potential biomarker for predicting breast cancer patients at increased risk for developing bone metastasis. In mouse models, IL-1B and its receptor (IL-1R1) are upregulated in breast cancer cells that metastasise to bone compared with cells that do not. We have now investigated the functional role of IL-1 by blocking IL-1R signalling with the clinically licensed antagonist, anakinra. Methodology 6-week old female BALB/c mice received a subcutaneous or intra-venous injection of MDA-MB-231-IV or MCF7 cells. Anakinra (1mg/kg/day) or placebo was administered 3 days before (preventative) or 7 days later (treatment). Tumour volume, apoptosis (TUNEL, Caspase 3), proliferation (Ki67) and angiogenesis (CD34, VEGF and endothelin) were analysed. Effects on bone were measured by uCT, and TRAP, P1NP, IL-1B, TNF alpha and IL-6 ELISA. Results Anakinra significantly reduced growth of MDA-MB-231-IV tumours in bone from 6.50+/3.00mm2 (placebo) to 2.56+/−1.07mm2 (treatment) and 0.63+/−0.18mm2 (preventative). Anakinra also reduced the number of mice that developed bone metastasis from 90% (placebo) to 40% (treatment) and 10% (preventative). Anti-tumour effects were not confined to bone, subcutaneous tumour volumes reduced from 656.68mm3 (placebo) to 160.47mm3 (treatment) and 31.08mm3 (preventative). Anakinra did not increase tumour cell apoptosis but reduced proliferation and angiogenesis in addition to exerting significant effects on the tumour environment reducing bone turnover markers, IL-1B and TNF alpha. Conclusions Our novel data demonstrate a functional role of IL-1 signalling in breast tumour progression and metastasis, supporting that anakinra could be repurposed for the treatment of breast cancer bone metastasis.
Collapse
Affiliation(s)
- Ingunn Holen
- Academic Unit of Clinical Oncology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield S10 2RX, UK
| | - Diane V Lefley
- Academic Unit of Clinical Oncology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield S10 2RX, UK
| | - Sheila E Francis
- Department of Infection Immunity and Cardiovascular Disease, University of Sheffield, Sheffield S10 2RX, UK
| | - Sarah Rennicks
- Academic Unit of Clinical Oncology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield S10 2RX, UK
| | - Steven Bradbury
- Academic Unit of Clinical Oncology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield S10 2RX, UK
| | - Robert E Coleman
- Academic Unit of Clinical Oncology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield S10 2RX, UK
| | - Penelope Ottewell
- Academic Unit of Clinical Oncology, Department of Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield S10 2RX, UK
| |
Collapse
|
12
|
Benyettou F, Alhashimi M, O'Connor M, Pasricha R, Brandel J, Traboulsi H, Mazher J, Olsen JC, Trabolsi A. Sequential Delivery of Doxorubicin and Zoledronic Acid to Breast Cancer Cells by CB[7]-Modified Iron Oxide Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2017; 9:40006-40016. [PMID: 29035507 DOI: 10.1021/acsami.7b11423] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Drug-loaded magnetic nanoparticles were synthesized and used for the sequential delivery of the antiresorptive agent zoledronic acid (Zol) and the cytotoxic drug doxorubicin (Dox) to breast cancer cells (MCF-7). Zol was attached to bare iron oxide nanoparticles (IONPs) via phosphonate coordination to form Z-NPs. The unbound imidazole of Zol was then used to complex the organic macrocycle CB[7] to obtain CZ-NPs. Dox was complexed to the CZ-NPs to form the fully loaded particles (DCZ-NPs), which were stable in solution at 37 °C and physiological pH (7.4). Fluorescence spectroscopy established that Dox is released in solution from DCZ-NPs suddenly (i) when the particles are subjected to magnetically induced heating to 42 °C at low pH (5.0) and (ii) in the presence of glutathione (GSH). Mass spectrometry indicated that Zol is released slowly in solution at low pH after Dox release. Magnetic measurements with a magnetic reader revealed that DCZ-NPs are internalized preferentially by MCF-7 cells versus nonmalignant cells (HEK293). Zol and Dox acted synergistically when delivered by the particles. DCZ-NPs caused a decrease in the viability of MCF-7 cells that was greater than the net decrease caused when the drugs were added to the cells individually at concentrations equivalent to those delivered by the particles. MCF-7 cells were treated with DCZ-NPs and subjected to an alternating magnetic field (AMF) which, with the nanoparticles present, raised the temperature of the cells and triggered the intracellular release of Dox, as indicated by fluorescence activated cell sorting (FACS). The cytotoxic effects of the DCZ-NPs on MCF-7 cells were enhanced 10-fold by AMF-induced heating. DCZ-NPs were also able to completely inhibit MCF-7 cell adhesion and invasion in vitro, indicating the potential of the particles to act as antimetastatic agents. Together these results demonstrate that DCZ-NPs warrant development as a system for combined chemo- and thermo-therapeutic treatment of cancer.
Collapse
Affiliation(s)
- Farah Benyettou
- New York University Abu Dhabi , P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Marwa Alhashimi
- New York University Abu Dhabi , P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Matthew O'Connor
- New York University Abu Dhabi , P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Renu Pasricha
- New York University Abu Dhabi , P.O. Box 129188, Abu Dhabi, United Arab Emirates
| | - Jeremy Brandel
- Equipe Reconnaissance et Procédés de Séparation Moléculaire, Université de Strasbourg , 67037 Strasbourg, France
| | - Hassan Traboulsi
- Chemistry Department, College of Sciences, King Faisal University-Al Ahsa , Hofuf 31982, Kingdom of Saudi Arabia
| | - Javed Mazher
- Physics Department, College of Sciences, King Faisal University-Al Ahsa , Hofuf 31982, Kingdom of Saudi Arabia
| | - John-Carl Olsen
- Department of Chemistry, University of Rochester RC 27021 , Rochester, New York 14607-0216, United States
| | - Ali Trabolsi
- New York University Abu Dhabi , P.O. Box 129188, Abu Dhabi, United Arab Emirates
| |
Collapse
|
13
|
Angeloni V, Contessi N, De Marco C, Bertoldi S, Tanzi MC, Daidone MG, Farè S. Polyurethane foam scaffold as in vitro model for breast cancer bone metastasis. Acta Biomater 2017; 63:306-316. [PMID: 28927931 DOI: 10.1016/j.actbio.2017.09.017] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 08/14/2017] [Accepted: 09/13/2017] [Indexed: 10/18/2022]
Abstract
Breast cancer (BC) represents the most incident cancer case in women (29%), with high mortality rate. Bone metastasis occurs in 20-50% cases and, despite advances in BC research, the interactions between tumor cells and the metastatic microenvironment are still poorly understood. In vitro 3D models gained great interest in cancer research, thanks to the reproducibility, the 3D spatial cues and associated low costs, compared to in vivo and 2D in vitro models. In this study, we investigated the suitability of a poly-ether-urethane (PU) foam as 3D in vitro model to study the interactions between BC tumor-initiating cells and the bone microenvironment. PU foam open porosity (>70%) appeared suitable to mimic trabecular bone structure. The PU foam showed good mechanical properties under cyclic compression (E=69-109kPa), even if lower than human trabecular bone. The scaffold supported osteoblast SAOS-2 cell line proliferation, with no cytotoxic effects. Human adipose derived stem cells (ADSC) were cultured and differentiated into osteoblast lineage on the PU foam, as shown by alizarin red staining and RT-PCR, thus offering a bone biomimetic microenvironment to the further co-culture with BC derived tumor-initiating cells (MCFS). Tumor aggregates were observed after three weeks of co-culture by e-cadherin staining and SEM; modification in CaP distribution was identified by SEM-EDX and associated to the presence of tumor cells. In conclusion, we demonstrated the suitability of the PU foam to reproduce a bone biomimetic microenvironment, useful for the co-culture of human osteoblasts/BC tumor-initiating cells and to investigate their interaction. STATEMENT OF SIGNIFICANCE 3D in vitro models represent an outstanding alternative in the study of tumor metastases development, compared to traditional 2D in vitro cultures, which oversimplify the 3D tissue microenvironment, and in vivo studies, affected by low reproducibility and ethical issues. Several scaffold-based 3D in vitro models have been proposed to recapitulate the development of metastases in different body sites but, still, the crucial challenge is to correctly mimic the tissue to be modelled in terms of physical, mechanical and biological properties. Here, we prove the suitability of a porous polyurethane foam, synthesized using an appropriate formulaton, in mimicking the bone tissue microenvironment and in reproducing the metastatic colonization derived from human breast cancer, particularly evidencing the devastating effects on the bone extracellular matrix caused by metastatic spreading.
Collapse
|
14
|
Baklaushev VP, Grinenko NF, Yusubalieva GM, Gubskii IL, Burenkov MS, Rabinovich EZ, Ivanova NV, Chekhonin VP. Mono- and Combined Therapy of Metastasizing Breast Carcinoma 4T1 with Zoledronic Acid and Doxorubicin. Bull Exp Biol Med 2016; 161:580-6. [PMID: 27590765 DOI: 10.1007/s10517-016-3464-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Indexed: 10/21/2022]
Abstract
The efficiency of monotherapy with zoledronic acid (Resorba), doxorubicin, and their combination was studied on the model of metastasizing breast carcinoma in BALB/c mice. Doxorubicin monotherapy was accompanied by a significant increase in median survival up to 57 days (vs. 34 and 35 days in control groups); 27% animals survived for 90 days (duration of the study). Bioluminescence area of the primary tumor significantly decreased on days 21 and 28; the total number of visceral metastases also decreased according to magnetic-resonance imaging data. Resorba monotherapy produced no general toxic effect, the median survival increased to 64 days, and 90-day survival was 33%. Imaging techniques (magnetic-resonance imaging, microtomography, bioluminescent analysis) showed that Resorba delayed the development of the primary tumor (regression of luminescence area on days 21 and 28, regression of standardized bioluminescence intensity on day 28) and significantly reduced the number of visceral metastases in comparison with the control. Combination therapy was less effective than monotherapy with the same medications. Median survival was 55 days, 90-day survival was 13%, but magnetic-resonance imaging and bioluminescence analysis after combination therapy also showed delayed growth of the primary tumor and reduced number of visceral metastases. Microtomography revealed bone metastases in ~30% animals of the control group; in experimental groups, no bone metastases were found. The experiment with periosteal (distal epiphysis of the femur) injection of 4T1-Luc2 tumor cells demonstrated pronounced selective effectiveness of Resorba in relation to bone metastases. Monotherapy with Resorba can prevent the development of not only bone, but also visceral metastases of breast cancer.
Collapse
Affiliation(s)
- V P Baklaushev
- Research and Educational Center of Medical Nanobiotechnologies, N. I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia. .,Federal Research Center of Specialized Health Care and Medical Technologies, Federal Medical-Biological Agency, Moscow, Russia.
| | - N F Grinenko
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky Federal Medical Research Centre for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - G M Yusubalieva
- Department of Fundamental and Applied Neurobiology, V. P. Serbsky Federal Medical Research Centre for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - I L Gubskii
- Research and Educational Center of Medical Nanobiotechnologies, N. I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | | | | | - N V Ivanova
- Research and Educational Center of Medical Nanobiotechnologies, N. I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia
| | - V P Chekhonin
- Research and Educational Center of Medical Nanobiotechnologies, N. I. Pirogov Russian National Research Medical University, Ministry of Health of the Russian Federation, Moscow, Russia.,Department of Fundamental and Applied Neurobiology, V. P. Serbsky Federal Medical Research Centre for Psychiatry and Narcology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
15
|
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
|
16
|
Yardley DA. Pharmacologic management of bone-related complications and bone metastases in postmenopausal women with hormone receptor-positive breast cancer. BREAST CANCER-TARGETS AND THERAPY 2016; 8:73-82. [PMID: 27217795 PMCID: PMC4861000 DOI: 10.2147/bctt.s97963] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
There is a high risk for bone loss and skeletal-related events, including bone metastases, in postmenopausal women with hormone receptor-positive breast cancer. Both the disease itself and its therapeutic treatments can negatively impact bone, resulting in decreases in bone mineral density and increases in bone loss. These negative effects on the bone can significantly impact morbidity and mortality. Effective management and minimization of bone-related complications in postmenopausal women with hormone receptor-positive breast cancer remain essential. This review discusses the current understanding of molecular and biological mechanisms involved in bone turnover and metastases, increased risk for bone-related complications from breast cancer and breast cancer therapy, and current and emerging treatment strategies for managing bone metastases and bone turnover in postmenopausal women with hormone receptor-positive breast cancer.
Collapse
Affiliation(s)
- Denise A Yardley
- Sarah Cannon Research Institute, Nashville, TN, USA; Tennessee Oncology, Nashville, TN, USA
| |
Collapse
|
17
|
Holen I, Walker M, Nutter F, Fowles A, Evans CA, Eaton CL, Ottewell PD. Oestrogen receptor positive breast cancer metastasis to bone: inhibition by targeting the bone microenvironment in vivo. Clin Exp Metastasis 2015; 33:211-24. [PMID: 26585891 DOI: 10.1007/s10585-015-9770-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 11/12/2015] [Indexed: 01/09/2023]
Abstract
Clinical trials have shown that adjuvant Zoledronic acid (ZOL) reduces the development of bone metastases irrespective of ER status. However, post-menopausal patients show anti-tumour benefit with ZOL whereas pre-menopausal patients do not. Here we have developed in vivo models of spontaneous ER+ve breast cancer metastasis to bone and investigated the effects of ZOL and oestrogen on tumour cell dissemination and growth. ER+ve (MCF7, T47D) or ER-ve (MDA-MB-231) cells were administered by inter-mammary or inter-cardiac injection into female nude mice ± estradiol. Mice were administered saline or 100 μg/kg ZOL weekly. Tumour growth, dissemination of tumour cells in blood, bone and bone turnover were monitored by luciferase imaging, histology, flow cytometry, two-photon microscopy, micro-CT and TRAP/P1NP ELISA. Estradiol induced metastasis of ER+ve cells to bone in 80-100 % of animals whereas bone metastases from ER-ve cells were unaffected. Administration of ZOL had no effect on tumour growth in the fat pad but significantly inhibited dissemination of ER+ve tumour cells to bone and frequency of bone metastasis. Estradiol and ZOL increased bone volume via different mechanisms: Estradiol increased activity of bone forming osteoblasts whereas administration of ZOL to estradiol supplemented mice decreased osteoclast activity and returned osteoblast activity to levels comparable to that of saline treated mice. ER-ve cells require increased osteoclast activity to grow in bone whereas ER+ve cells do not. Zol does not affect ER+ve tumour growth in soft tissue, however, inhibition of bone turnover by ZOL reduced dissemination and growth of ER+ve breast cancer cells in bone.
Collapse
Affiliation(s)
- I Holen
- Academic Unit of Clinical Oncology, Department of Oncology, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - M Walker
- Academic Unit of Clinical Oncology, Department of Oncology, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - F Nutter
- Academic Unit of Clinical Oncology, Department of Oncology, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - A Fowles
- Bone Biology, Department of Human Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, S10 2RX, UK
| | - C A Evans
- Academic Unit of Clinical Oncology, Department of Oncology, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - C L Eaton
- Bone Biology, Department of Human Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, S10 2RX, UK
| | - P D Ottewell
- Academic Unit of Clinical Oncology, Department of Oncology, Medical School, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK.
| |
Collapse
|
18
|
Ye WL, Zhao YP, Na R, Li F, Mei QB, Zhao MG, Zhou SY. Actively Targeted Delivery of Doxorubicin to Bone Metastases by a pH-Sensitive Conjugation. J Pharm Sci 2015; 104:2293-303. [DOI: 10.1002/jps.24476] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 03/24/2015] [Accepted: 04/16/2015] [Indexed: 11/08/2022]
|
19
|
Ottewell PD, Wang N, Brown HK, Fowles CA, Croucher PI, Eaton CL, Holen I. OPG-Fc inhibits ovariectomy-induced growth of disseminated breast cancer cells in bone. Int J Cancer 2015; 137:968-77. [PMID: 25603921 DOI: 10.1002/ijc.29439] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 12/29/2014] [Indexed: 01/28/2023]
Abstract
Dormant disseminated tumour cells can be detected in the bone marrow of breast cancer patients several years after resection of the primary tumour. The majority of these patients will remain asymptomatic, however, ∼ 15% will go on to develop overt bone metastases and this condition is currently incurable. The reason why these dormant cells are stimulated to proliferate and form bone tumours in some patients and not others remains to be elucidated. We have recently shown that in an in vivo model, increasing bone turnover by ovariectomy stimulated proliferation of disseminated tumour cells, resulting in formation of bone metastasis. We now show for the first time that osteoclast mediated mechanisms induce growth of tumours from dormant MDA-MB-231 cells disseminated in the bone. We also show that disruption of RANK-RANKL interactions following administration of OPG-Fc inhibits growth of these dormant tumour cells in vivo. Our data support early intervention with anti-resorptive therapy in a low-oestrogen environment to prevent development of bone metastases.
Collapse
Affiliation(s)
- Penelope D Ottewell
- Department of Oncology, Academic Unit of Clinical Oncology, University of Sheffield, S10 2RX, United Kingdom
| | - Ning Wang
- Academic Unit of Bone Biology, Department of Human Metabolism, University of Sheffield, S10 2RX, United Kingdom
| | - Hannah K Brown
- Department of Oncology, Academic Unit of Clinical Oncology, University of Sheffield, S10 2RX, United Kingdom
| | - C Anne Fowles
- Academic Unit of Bone Biology, Department of Human Metabolism, University of Sheffield, S10 2RX, United Kingdom
| | - Peter I Croucher
- Musculoskeletal Medicine Division, Garvan Institute of Medical Research, Sidney, New South Wales, Australia
| | - Colby L Eaton
- Academic Unit of Bone Biology, Department of Human Metabolism, University of Sheffield, S10 2RX, United Kingdom
| | - Ingunn Holen
- Department of Oncology, Academic Unit of Clinical Oncology, University of Sheffield, S10 2RX, United Kingdom
| |
Collapse
|
20
|
Ahn SG, Kim SH, Lee HM, Lee SA, Jeong J. Survival benefit of zoledronic Acid in postmenopausal breast cancer patients receiving aromatase inhibitors. J Breast Cancer 2014; 17:350-5. [PMID: 25548583 PMCID: PMC4278054 DOI: 10.4048/jbc.2014.17.4.350] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/30/2014] [Indexed: 11/30/2022] Open
Abstract
Purpose A growing body of evidence indicates that zoledronic acid (ZA) can improve the clinical outcome in patients with breast cancer and low estrogen levels. In the present study, we aimed to investigate the survival benefit of ZA administration in postmenopausal Korean women with breast cancer who were also receiving aromatase inhibitors. Methods Between January 2004 and December 2010, 235 postmenopausal breast cancer patients undergoing aromatase inhibitor therapy were investigated. All patients were postmenopausal, as confirmed by laboratory tests. Of these patients, 77 received adjuvant upfront ZA for at least 1 year in addition to conventional adjuvant treatment. The remaining 158 patients never received ZA and were treated according to the St. Gallen guidelines. Results The baseline characteristics for ZA treatment were not different between the two groups. The median follow-up time was 62 months, and the patients who received ZA in addition to aromatase inhibitors showed a better recurrence-free survival compared to those who received aromatase inhibitors alone (p=0.035). On multivariate analysis, the patients who received ZA showed a better recurrence-free survival independent of the tumor size, nodal status, progesterone receptor, and histological grade. For this model, Harrell c index was 0.743. The hazard ratio of ZA use for recurrence-free survival was 0.12 (95% confidence interval, 0.01-0.99). Conclusion Our findings suggest that upfront use of ZA as part of adjuvant treatment can offer a survival benefit to postmenopausal breast cancer patients receiving aromatase inhibitor treatment.
Collapse
Affiliation(s)
- Sung Gwe Ahn
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Sung Hyun Kim
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Hak Min Lee
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| | - Seung Ah Lee
- Department of Surgery, Eulji University College of Medicine, Seoul, Korea
| | - Joon Jeong
- Department of Surgery, Yonsei University College of Medicine, Seoul, Korea
| |
Collapse
|
21
|
Lipton A. Zoledronic acid: multiplicity of use across the cancer continuum. Expert Rev Anticancer Ther 2014; 11:999-1012. [DOI: 10.1586/era.11.71] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
22
|
Daniele G, Giordano P, De Luca A, Piccirillo MC, Di Maio M, Giudice AD, De Feo G, Bryce J, Lamura L, Vecchione A, Normanno N, Perrone F. Anticancer effect of bisphosphonates: new insights from clinical trials and preclinical evidence. Expert Rev Anticancer Ther 2014; 11:299-307. [DOI: 10.1586/era.10.214] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
23
|
Charehbili A, Fontein DBY, Kroep JR, Liefers GJ, Nortier JWR, Velde CJHVD. Can Zoledronic Acid be Beneficial for Promoting Tumor Response in Breast Cancer Patients Treated with Neoadjuvant Chemotherapy? J Clin Med 2013; 2:188-200. [PMID: 26237143 PMCID: PMC4470143 DOI: 10.3390/jcm2040188] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Revised: 09/05/2013] [Accepted: 09/11/2013] [Indexed: 01/27/2023] Open
Abstract
The antitumor effect of bisphosphonates (BPs) is under increasing scrutiny. Preclinical and clinical evidence has shown that BPs might sensitize breast tumors to chemotherapy. Here, we present a review of current preclinical and clinical evidence for antitumor effects of BPs, and evaluate how BPs might play a role in neoadjuvant treatment of women with breast cancer.
Collapse
Affiliation(s)
- Ayoub Charehbili
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, ZA Leiden 2333, The Netherlands.
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, ZA Leiden 2333, The Netherlands.
| | - Duveken B Y Fontein
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, ZA Leiden 2333, The Netherlands.
| | - Judith R Kroep
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, ZA Leiden 2333, The Netherlands.
| | - Gerrit-Jan Liefers
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, ZA Leiden 2333, The Netherlands.
| | - Johannes W R Nortier
- Department of Medical Oncology, Leiden University Medical Center, Albinusdreef 2, ZA Leiden 2333, The Netherlands.
| | - Cornelis J H van de Velde
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, ZA Leiden 2333, The Netherlands.
| |
Collapse
|
24
|
De Rosa G, Misso G, Salzano G, Caraglia M. Bisphosphonates and cancer: what opportunities from nanotechnology? JOURNAL OF DRUG DELIVERY 2013; 2013:637976. [PMID: 23533771 PMCID: PMC3603225 DOI: 10.1155/2013/637976] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 01/22/2013] [Indexed: 02/04/2023]
Abstract
Bisphosphonates (BPs) are synthetic analogues of naturally occurring pyrophosphate compounds. They are used in clinical practice to inhibit bone resorption in bone metastases, osteoporosis, and Paget's disease. BPs induce apoptosis because they can be metabolically incorporated into nonhydrolyzable analogues of adenosine triphosphate. In addition, the nitrogen-containing BPs (N-BPs), second-generation BPs, act by inhibiting farnesyl diphosphate (FPP) synthase, a key enzyme of the mevalonate pathway. These molecules are able to induce apoptosis of a number of cancer cells in vitro. Moreover, antiangiogenic effect of BPs has also been reported. However, despite these promising properties, BPs rapidly accumulate into the bone, thus hampering their use to treat extraskeletal tumors. Nanotechnologies can represent an opportunity to limit BP accumulation into the bone, thus increasing drug level in extraskeletal sites of the body. Thus, nanocarriers encapsulating BPs can be used to target macrophages, to reduce angiogenesis, and to directly kill cancer cell. Moreover, nanocarriers can be conjugated with BPs to specifically deliver anticancer agent to bone tumors. This paper describes, in the first part, the state-of-art on the BPs, and, in the following part, the main studies in which nanotechnologies have been proposed to investigate new indications for BPs in cancer therapy.
Collapse
Affiliation(s)
- Giuseppe De Rosa
- Department of Pharmacy, Università degli Studi di Napoli Federico II, Via Domenico Montesano 49, 8013 Naples, Italy
| | - Gabriella Misso
- Department of Biochemistry, Biophysics and General Pathology, Seconda Università degli Studi di Napoli, Via Costantinopoli 16, 80138 Naples, Italy
| | - Giuseppina Salzano
- Department of Pharmacy, Università degli Studi di Napoli Federico II, Via Domenico Montesano 49, 8013 Naples, Italy
| | - Michele Caraglia
- Department of Biochemistry, Biophysics and General Pathology, Seconda Università degli Studi di Napoli, Via Costantinopoli 16, 80138 Naples, Italy
| |
Collapse
|
25
|
Ibrahim T, Liverani C, Mercatali L, Sacanna E, Zanoni M, Fabbri F, Zoli W, Amadori D. Cisplatin in combination with zoledronic acid: a synergistic effect in triple-negative breast cancer cell lines. Int J Oncol 2013; 42:1263-70. [PMID: 23403907 DOI: 10.3892/ijo.2013.1809] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 11/20/2012] [Indexed: 11/06/2022] Open
Abstract
Zoledronic acid (ZA) is the most widely used bisphos-phonate to treat cancer-induced bone disease. There is evidence that bisphosphonates have direct antitumor activity and that their combination with anticancer agents can significantly enhance the effect of treatment. We evaluated whether the combination of ZA with different platinum compounds exerts a synergistic effect in breast cancer cell lines and we investigated the mechanisms of action involved. This study was performed on four breast cancer cell lines, MCF-7, SKBR3, MDA-MB-231 and BRC-230, and confirmed on a primary culture obtained from a breast cancer bone metastasis specimen. ZA (50 µM) was administered for 72 h alone or in combination with cisplatin (Cis) or carboplatin. Drug-induced growth inhibition was detected by sulforhodamine B assay, apoptosis and cell cycle regulation were detected by flow cytometry, and protein expression was evaluated by western blot analysis. MCF-7 and SKBR3 showed very low sensitivity to the three drugs tested. The ZA + Cis combination exerted a high antitumor activity in the two triple-negative lines MDA-MB-231 and BRC-230. An important synergistic effect was obtained in MDA-MB-231 and an additive effect was observed in BRC-230. The p21, pMAPK and m-TOR pathways were regulated by this combined treatment, particularly at lower Cis doses. Carboplatin did not show antitumor activity either alone or in combination with ZA. In conclusion, the potential novel treatment schedule identified for triple-negative breast cancer could prove beneficial in view of the limited therapeutic options available for patients and also since the synergism with ZA would enable lower Cis doses to be used, thus reducing toxicity. Although further research in a clinical setting is warranted, our results on cell lines has been confirmed on a human primary bone metastasis culture.
Collapse
Affiliation(s)
- Toni Ibrahim
- Osteoncology Center, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (I.R.S.T.), I-47014 Meldola (FC), Italy.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Adjuvant bone-targeted therapy to prevent metastasis: lessons from the AZURE study. Curr Opin Support Palliat Care 2013; 6:322-9. [PMID: 22801464 DOI: 10.1097/spc.0b013e32835689cd] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW Bone-targeted treatments with bisphosphonates (e.g., zoledronic acid) and denosumab are known to reduce the risk of skeletal complications and prevent treatment-induced bone loss in patients with malignant bone disease. Additionally, these drugs may modify the course of bone destruction via inhibitory effects on the 'vicious cycle' of growth factor and cytokine signalling between tumour and bone cells within the bone marrow microenvironment. RECENT FINDINGS In early stage breast cancer, treatment with zoledronic acid has shown improvements in disease-free and overall survival, notably in women with established menopause at diagnosis and in premenopausal women with endocrine-responsive disease, treated with goserelin to suppress ovarian function. Other bisphosphonates such as clodronate may produce similar benefits. Additionally, in castrate-resistant prostate cancer, treatment with denosumab delays the development of bone metastases. SUMMARY These results strongly support the adjuvant use of bone-targeted treatments, but suggest that reproductive hormones are an important treatment modifier to take into account.
Collapse
|
27
|
Ibrahim T, Mercatali L, Sacanna E, Tesei A, Carloni S, Ulivi P, Liverani C, Fabbri F, Zanoni M, Zoli W, Amadori D. Inhibition of breast cancer cell proliferation in repeated and non-repeated treatment with zoledronic acid. Cancer Cell Int 2012; 12:48. [PMID: 23173568 PMCID: PMC3519531 DOI: 10.1186/1475-2867-12-48] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 11/14/2012] [Indexed: 01/25/2023] Open
Abstract
Background Zoledronic acid is used to treat bone metastases and has been shown to reduce skeletal-related events and exert antitumor activity. The present in vitro study investigates the mechanism of action of Zoledronic Acid on breast cancer cell lines with different hormonal and HER2 patterns. Furthermore, we investigated the efficacy of repeated versus non-repeated treatments. Methods The study was performed on 4 breast cancer cell lines (BRC-230, SkBr3, MCF-7 and MDA-MB-231). Non-repeated treatment (single exposure of 168 hrs’ duration) with zoledronic acid was compared with repeated treatment (separate exposures, each of 48 hrs’ duration, for a total of 168 hrs) at different dosages. A dose–response profile was generated using sulforhodamine B assay. Apoptosis was evaluated by TUNEL assay and biomolecular characteristics were analyzed by western blot. Results Zoledronic acid produced a dose-dependent inhibition of proliferation in all cell lines. Anti-proliferative activity was enhanced with the repeated treatment, proving to be statistically significant in the triple-negative lines. In these lines repeated treatment showed a cytocidal effect, with apoptotic cell death caused by caspase 3, 8 and 9 activation and decreased RAS and pMAPK expression. Apoptosis was not observed in estrogen receptor-positive line: p21 overexpression suggested a slowing down of cell cycle. A decrease in RAS and pMAPK expression was seen in HER2-overexpressing line after treatment. Conclusions The study suggests that zoledronic acid has an antitumor activity in breast cancer cell lines. Its mechanism of action involves the decrease of RAS and RHO, as in osteoclasts. Repeated treatment enhances antitumor activity compared to non-repeated treatment. Repeated treatment has a killing effect on triple-negative lines due to apoptosis activation. Further research is warranted especially in the treatment of triple-negative breast cancer.
Collapse
Affiliation(s)
- Toni Ibrahim
- Osteoncology Center, IRCCS Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), via P, Maroncelli 40, 47014, Meldola, FC, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Hadji P, Coleman R, Gnant M, Green J. The impact of menopause on bone, zoledronic acid, and implications for breast cancer growth and metastasis. Ann Oncol 2012; 23:2782-2790. [PMID: 22730099 PMCID: PMC3477882 DOI: 10.1093/annonc/mds169] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Revised: 04/02/2012] [Accepted: 04/17/2012] [Indexed: 12/22/2022] Open
Abstract
Recent data from the AZURE, ABCSG-12, and ZO-FAST clinical trials have challenged our understanding of the potential anticancer activity of zoledronic acid (ZOL). Although the results of these studies may appear to be conflicting on the surface, a deeper look into commonalities among the patient populations suggest that some host factors (i.e. patient age and endocrine status) may contribute to the anticancer activity of ZOL. Indeed, data from these large clinical trials suggest that the potential anticancer activity of ZOL may be most robust in a low-estrogen environment. However, this may be only part of the story and many questions remain to be answered to fully explain the phenomenon. Does estrogen override the anticancer activity of ZOL seen in postmenopausal women? Are hormones other than estrogen involved that contribute to this effect? Does the role of bone turnover in breast cancer (BC) growth and progression differ in the presence of various estrogen levels? Here, we present a review of the multitude of factors affected by different endocrine environments in women with BC that may influence the potential anticancer activity of ZOL.
Collapse
Affiliation(s)
- P Hadji
- Department of Gynecology, Endocrinology, and Oncology, Philipps-University of Marburg, Marburg, Germany
| | - R Coleman
- Academic Unit of Clinical Oncology, Weston Park Hospital, CR-UK/YCR Sheffield Cancer Research Centre, Sheffield, UK
| | - M Gnant
- Department of Surgery, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - J Green
- Department of Oncology, Novartis Pharma AG, Basel, Switzerland.
| |
Collapse
|
29
|
Brown HK, Ottewell PD, Evans CA, Coleman RE, Holen I. A single administration of combination therapy inhibits breast tumour progression in bone and modifies both osteoblasts and osteoclasts. J Bone Oncol 2012; 1:47-56. [PMID: 26909255 PMCID: PMC4723328 DOI: 10.1016/j.jbo.2012.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 05/16/2012] [Accepted: 05/22/2012] [Indexed: 12/11/2022] Open
Abstract
We have previously shown that repeated sequential administration of doxorubicin, followed 24 h later by zoledronic acid, inhibits tumour growth in models of established breast cancer bone metastasis. As breast cancer patients only receive zoledronic acid every 3–4 weeks, the aim of the current study was to establish the anti-tumour and bone effects of a single administration of doxorubicin/zoledronic acid combination therapy in a bone metastasis model. MDA-MB-231-GFP cells were injected i.c. in 6-week-old nude mice. On day 2, animals received PBS, doxorubicin (2 mg/kg i.v.), zoledronic acid (100 μg/kg s.c.) or doxorubicin followed 24 h later by zoledronic acid. Anti-tumour effects were assessed on days 15/23 by quantification of apoptotic and proliferating cells and changes in expression of genes implicated in apoptosis, proliferation and bone turnover. Bone effects were assessed by μCT analysis, bone histomorphometry and measurement of serum markers. A tumour-free control group was included. Combination treatment reduced bone tumour burden compared to single agent or PBS control and increased levels of tumour cell apoptosis on day 15, but this was no longer detectable on day 23. Animals receiving zoledronic acid had increased bone density, without evidence of tumour-induced lesions. Bone histomorphometry showed that zoledronic acid caused a decrease in osteoblast and osteoclast numbers and an increase in osteoclast size, in both tumour-free and tumour-bearing animals. Our data show that although zoledronic acid modifies the bone microenvironment through effects on both osteoblasts and osteoclasts, this does not result in a significant anti-tumour effect in the absence of doxorubicin.
Collapse
Affiliation(s)
- H K Brown
- Academic Unit of Clinical Oncology, CR-UK/YCR Sheffield Cancer Research Centre, University of Sheffield, UK
| | - P D Ottewell
- Academic Unit of Clinical Oncology, CR-UK/YCR Sheffield Cancer Research Centre, University of Sheffield, UK
| | - C A Evans
- Academic Unit of Clinical Oncology, CR-UK/YCR Sheffield Cancer Research Centre, University of Sheffield, UK
| | - R E Coleman
- Academic Unit of Clinical Oncology, CR-UK/YCR Sheffield Cancer Research Centre, University of Sheffield, UK
| | - I Holen
- Academic Unit of Clinical Oncology, CR-UK/YCR Sheffield Cancer Research Centre, University of Sheffield, UK
| |
Collapse
|
30
|
Li XY, Lin YC, Huang WL, Hong CQ, Chen JY, You YJ, Li WB. Zoledronic acid inhibits proliferation and impairs migration and invasion through downregulating VEGF and MMPs expression in human nasopharyngeal carcinoma cells. Med Oncol 2012; 29:714-20. [PMID: 21431960 DOI: 10.1007/s12032-011-9904-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Accepted: 03/08/2011] [Indexed: 02/05/2023]
Abstract
To demonstrate the effect of zoledronic acid in proliferation, invasion, and migration of human nasopharyngeal carcinoma cell HNE-1 and explore the potential role of VEGF, MMP-2, and MMP-9 proteins in vitro. Human nasopharyngeal carcinoma cell HNE-1 was exposed to various concentrations (0-40 μmol/l) of zoledronic acid. Zoledronic acid inhibited proliferation of HNE-1 cells though not in a dose-dependent manner. Zoledronic acid had exerted a dose-dependent effect on the migration and invasion of HNE-1 cells. Both expressions of mRNA and protein of MMP2, MMP9, and VEGF were reduced, respectively, detected by RT-PCR and Western blot assays. These data suggested that zoledronic acid not only inhibited growth but also invasion and migration of HNE-1 cells in vitro. The anti-cancer action of zoledronic acid was partially associated with the suppression of VEGF expression and secretion and downregulating the expression of MMP2 and MMP9.
Collapse
Affiliation(s)
- Xu-Yuan Li
- Department of Internal Medicine, Cancer Hospital of Shantou University Medical College, 7 Raoping Road, Shantou 515031, China
| | | | | | | | | | | | | |
Collapse
|
31
|
Holen I, Whitworth J, Nutter F, Evans A, Brown HK, Lefley DV, Barbaric I, Jones M, Ottewell PD. Loss of plakoglobin promotes decreased cell-cell contact, increased invasion, and breast cancer cell dissemination in vivo. Breast Cancer Res 2012; 14:R86. [PMID: 22632416 PMCID: PMC3446349 DOI: 10.1186/bcr3201] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2011] [Revised: 05/10/2012] [Accepted: 05/25/2012] [Indexed: 12/25/2022] Open
Abstract
Introduction The majority of deaths from breast cancer are a result of metastases; however, little is understood about the genetic alterations underlying their onset. Genetic profiling has identified the adhesion molecule plakoglobin as being three-fold reduced in expression in primary breast tumors that have metastasized compared with nonmetastatic tumors. In this study, we demonstrate a functional role for plakoglobin in the shedding of tumor cells from the primary site into the circulation. Methods We investigated the effects of plakoglobin knockdown on breast cancer cell proliferation, migration, adhesion, and invasion in vitro and on tumor growth and intravasation in vivo. MCF7 and T47D cells were stably transfected with miRNA sequences targeting the plakoglobin gene, or scramble vector. Gene and protein expression was monitored by quantitative polymerase chain reaction (qPCR) and Western blot. Cell proliferation, adhesion, migration, and invasion were measured by cell counting, flow cytometry, and scratch and Boyden Chamber assays. For in vivo experiments, plakoglobin knockdown and control cells were inoculated into mammary fat pads of mice, and tumor growth, shedding of tumor cells into the bloodstream, and evidence of metastatic bone lesions were monitored with caliper measurement, flow cytometry, and microcomputed tomography (μCT), respectively. Results Plakoglobin and γ-catenin expression were reduced by more than 80% in all knockdown cell lines used but were unaltered after transfection with the scrambled sequence. Reduced plakoglobin resulted in significantly increased in MCF7 and T47D cell proliferation in vitro and in vivo, compared with control, with significantly more tumor cells being shed into the bloodstream of mice bearing plakoglobin knockdown tumors. In addition, plakoglobin knockdown cells showed a >250% increase in invasion through basement membrane and exhibited reduced cell-to-cell adhesion compared with control cells. Conclusion Decreased plakoglobin expression increases the invasive behavior of breast cancer cells. This is the first demonstration of a functional role for plakoglobin/γ-catenin in the metastatic process, indicating that this molecule may represent a target for antimetastatic therapies.
Collapse
Affiliation(s)
- Ingunn Holen
- Academic Unit of Clinical Oncology, University of Sheffield, Medical School, Beech Hill Road, Sheffield, S10 2RX, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Russell K, Clemons M, Costa L, Addison CL. Adjuvant bisphosphonate treatment for breast cancer: Where are we heading and can the pre-clinical literature help us get there? J Bone Oncol 2012; 1:12-7. [PMID: 26909249 PMCID: PMC4723323 DOI: 10.1016/j.jbo.2012.04.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 04/14/2012] [Accepted: 04/16/2012] [Indexed: 11/18/2022] Open
Abstract
Bisphosphonates have demonstrated anti-tumour activity in preclinical studies of bone metastatic disease, thus it was natural to transition these agents into the adjuvant cancer therapy setting. Surprisingly, the results of adjuvant breast cancer trials have shown either modest to no benefit or even harm. We sought to explore whether the preclinical results supporting bisphosphonate use provided clues to help explain the current clinical data. Interestingly, the majority of preclinical data suggested that bisphosphonate treatment was more efficacious when administered after the establishment of osseous metastases. This is similar to the findings of one clinical study whereby patients with biopsy evidence of osseous micrometastases derive greater survival benefit from bisphosphonate treatment. Another clinical study found bisphosphonates were associated with increased incidence of visceral metastases, similar to what has been previously published in preclinical models using “preventative” dosing strategies. While the current clinical data suggest bisphosphonates may be more efficacious in post-menopausal or oestrogen depleted patients, or those with hormone receptor positive tumours, to date no appropriately designed preclinical studies have evaluated these effects. Furthermore, putative mechanisms that regulate response to bisphosphonates in other tumour types remain to be evaluated in breast cancer. Despite the initial optimism regarding adjuvant bisphosphonate therapy, the conflicting clinical results from large trials suggest that we should return to the bench to further investigate factors that may influence response to bisphosphonate treatment or identify appropriate characteristics that would indicate the sub-groups of patients most likely to benefit from bisphosphonate treatment.
Collapse
Affiliation(s)
- Kent Russell
- Division of Medical Oncology, Ottawa Hospital Cancer Centre and Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Mark Clemons
- Division of Medical Oncology, Ottawa Hospital Cancer Centre and Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Luis Costa
- Serviço de Oncologia do Hospital de Santa Maria and Instituto de Medicina Molecular, Lisbon, Portugal
| | - Christina L. Addison
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Corresponding author at: Cancer Therapeutics Program, Ottawa Hospital Research Institute, Box 926, 3rd Floor TOHRCC, Ottawa, ON, Canada K1H 8L6. Tel.: +1 613 737 7700; fax: +1 613 247 3524.
| |
Collapse
|
33
|
Gnant M. Zoledronic acid in the treatment of early-stage breast cancer: is there a final verdict? Curr Oncol Rep 2012; 14:35-43. [PMID: 22113793 DOI: 10.1007/s11912-011-0209-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
Breast cancer, which preferentially metastasizes to bone, is the most common malignancy among women worldwide and is a leading cause of death. Clinical data from large, phase 3 trials (ie, ABCSG-12, ZO-FAST, and AZURE) demonstrate significantly improved disease-free survival with zoledronic acid in some patient populations with early breast cancer. Although the interim results from the AZURE trial did not show a disease-free survival benefit with zoledronic acid in the overall patient population, subset analyses showed that zoledronic acid significantly improved disease-free survival in women with established postmenopausal status at baseline. Similarly, subset analyses of the ABCSG-12 trial showed greater benefits from zoledronic acid in patients over 40 years of age who theoretically would have achieved more complete ovarian suppression. Together, these data support a potential role for zoledronic acid beyond bone health in breast cancer and suggest that the endocrine environment may influence the anticancer potential of zoledronic acid.
Collapse
Affiliation(s)
- Michael Gnant
- Department of Surgery, Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, A-1090, Vienna, Austria.
| |
Collapse
|
34
|
Gallo M, De Luca A, Lamura L, Normanno N. Zoledronic acid blocks the interaction between mesenchymal stem cells and breast cancer cells: implications for adjuvant therapy of breast cancer. Ann Oncol 2012; 23:597-604. [PMID: 21551002 DOI: 10.1093/annonc/mdr159] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Zoledronic acid (ZA) reduces locoregional and distant metastases in estrogen receptor (ER)-positive breast cancer patients. Since ZA rapidly concentrates in the bone following i.v. administration, we hypothesized that this phenomenon involves the mechanism of action of ZA in bone tissue. MATERIALS AND METHODS Migration assays were carried out in fibronectin-coated Boyden chambers. Activation of signaling proteins was analyzed with a phosphoprotein array. Chemokines and growth factors were measured by immunoassays and real-time PCR. RESULTS ZA significantly reduced in bone marrow-derived mesenchymal stem cells (MSCs) the activation of AKT and mitogen-activated protein kinase and their ability to migrate. Conditioned medium (CM) from ZA-treated MSCs showed a reduced capacity to promote the migration of ER-positive MCF-7 breast cancer cells as compared with CM from untreated MSCs. The levels of the chemokine (C-C motif) ligand 5 (CCL5, also known as RANTES - Regulated upon Activation, Normal T-cell Expressed, and Secreted) and interleukin (IL)-6 were significantly reduced in MSC-CM following treatment with ZA. Anti-RANTES and anti-IL-6 antibodies almost completely abolished the migration of MCF-7 cells induced by MSC-CM. Recombinant RANTES and IL-6 significantly induced MCF-7 cell migration and their combination showed a cooperative effect. Similar results were observed in different breast cancer cell lines. CONCLUSION ZA might exert its antitumor activity by inhibiting MSC migration and blocking MSCs' secretion of factors involved in breast cancer progression.
Collapse
Affiliation(s)
- M Gallo
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - "Fondazione Pascale", Naples, Italy
| | - A De Luca
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - "Fondazione Pascale", Naples, Italy
| | - L Lamura
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - "Fondazione Pascale", Naples, Italy
| | - N Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - "Fondazione Pascale", Naples, Italy.
| |
Collapse
|
35
|
Michailidou M, Holen I. Combinations of bisphosphonates and classical anticancer drugs: a preclinical perspective. Recent Results Cancer Res 2012; 192:145-69. [PMID: 22307374 DOI: 10.1007/978-3-642-21892-7_7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Bone metastases are frequent complications in advanced breast and prostate cancer among others, resulting in increased risk of fractures, pain, hypercalcaemia of malignancy and a reduction in patient independence and mobility. Bisphosphonates (BPs) are in wide clinical use for the treatment of cancer-induced bone disease associated with advanced cancer, due to their potent ability to reduce skeletal-related events (SREs) and improve quality of life. Despite the profound effect on bone health, the majority of clinical studies have failed to demonstrate an overall survival benefit of BP therapy. There is increasing preclinical evidence to suggest that inclusion of the most potent nitrogen-containing BPs (NBPs) in combination therapy results in increased antitumour effects and improved survival, but that the particular schedules used are of key importance to achieve optimal benefit. Recent clinical data have suggested that there may be effects of adjuvant NBP therapy on breast tumours outside the skeleton. These findings have led to renewed interest in the use of BPs in cancer therapy, in particular how they can be included as part of adjuvant protocols. Here we review the key data reported from preclinical model systems investigating the effects of combination therapy including BPs with particular emphasis on breast and prostate cancer.
Collapse
|
36
|
Yan T, Yin W, Zhou Q, Zhou L, Jiang Y, Du Y, Shao Z, Lu J. The efficacy of zoledronic acid in breast cancer adjuvant therapy: A meta-analysis of randomised controlled trials. Eur J Cancer 2012; 48:187-95. [DOI: 10.1016/j.ejca.2011.10.021] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2011] [Revised: 08/30/2011] [Accepted: 10/17/2011] [Indexed: 11/28/2022]
|
37
|
Clinical considerations for the use of antiresorptive agents in the treatment of metastatic bone disease. Crit Rev Oncol Hematol 2011; 80:301-13. [DOI: 10.1016/j.critrevonc.2011.03.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Revised: 03/10/2011] [Accepted: 03/24/2011] [Indexed: 01/30/2023] Open
|
38
|
Coleman RE, Marshall H, Cameron D, Dodwell D, Burkinshaw R, Keane M, Gil M, Houston SJ, Grieve RJ, Barrett-Lee PJ, Ritchie D, Pugh J, Gaunt C, Rea U, Peterson J, Davies C, Hiley V, Gregory W, Bell R. Breast-cancer adjuvant therapy with zoledronic acid. N Engl J Med 2011; 365:1396-405. [PMID: 21995387 DOI: 10.1056/nejmoa1105195] [Citation(s) in RCA: 360] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Data suggest that the adjuvant use of bisphosphonates reduces rates of recurrence and death in patients with early-stage breast cancer. We conducted a study to determine whether treatment with zoledronic acid, in addition to standard adjuvant therapy, would improve disease outcomes in such patients. METHODS In this open-label phase 3 study, we randomly assigned 3360 patients to receive standard adjuvant systemic therapy either with or without zoledronic acid. The zoledronic acid was administered every 3 to 4 weeks for 6 doses and then every 3 to 6 months to complete 5 years of treatment. The primary end point of the study was disease-free survival. A second interim analysis revealed that a prespecified boundary for lack of benefit had been crossed. RESULTS At a median follow-up of 59 months, there was no significant between-group difference in the primary end point, with a rate of disease-free survival of 77% in each group (adjusted hazard ratio in the zoledronic acid group, 0.98; 95% confidence interval [CI], 0.85 to 1.13; P=0.79). Disease recurrence or death occurred in 377 patients in the zoledronic acid group and 375 of those in the control group. The numbers of deaths--243 in the zoledronic acid group and 276 in the control group--were also similar, resulting in rates of overall survival of 85.4% in the zoledronic acid group and 83.1% in the control group (adjusted hazard ratio, 0.85; 95% CI, 0.72 to 1.01; P=0.07). In the zoledronic acid group, there were 17 confirmed cases of osteonecrosis of the jaw (cumulative incidence, 1.1%; 95% CI, 0.6 to 1.7; P<0.001) and 9 suspected cases; there were no cases in the control group. Rates of other adverse effects were similar in the two study groups. CONCLUSIONS These findings do not support the routine use of zoledronic acid in the adjuvant management of breast cancer. (Funded by Novartis Pharmaceuticals and the National Cancer Research Network; AZURE Current Controlled Trials number, ISRCTN79831382.).
Collapse
Affiliation(s)
- Robert E Coleman
- Academic Unit of Clinical Oncology, Weston Park Hospital, University of Sheffield, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Gnant M, Clézardin P. Direct and indirect anticancer activity of bisphosphonates: a brief review of published literature. Cancer Treat Rev 2011; 38:407-15. [PMID: 21983264 DOI: 10.1016/j.ctrv.2011.09.003] [Citation(s) in RCA: 117] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 09/07/2011] [Accepted: 09/12/2011] [Indexed: 01/02/2023]
Abstract
The bone marrow microenvironment provides a site for cancer cells to evade systemic anticancer therapy. Dormant tumor micrometastases are believed to be the source of disease persistence and relapse; however, the exact characteristics of cancer stem cells vs. cancer cells with limited metastatic potential have yet to be elucidated. Bisphosphonates inhibit osteoclast-mediated bone resorption, are approved for treating malignant bone disease from advanced cancers, and have shown efficacy for preventing cancer treatment-induced bone loss. Altering the bone marrow microenvironment to make it less conducive to cancer cell survival is now emerging as an important means to prevent cancer recurrence. This review aims to distill the diverse literature and provide a brief overview of the numerous preclinical and early clinical studies of bisphosphonates demonstrating a variety of direct and indirect anticancer activities that affect both the tumor cell (the "seed") and surrounding microenvironment (the "soil"). Recently, zoledronic acid was found to improve disease-free survival and overall survival in some adjuvant breast cancer settings and prolonged survival in patients with multiple myeloma and other advanced cancers. In the prostate cancer setting, antiresorptive therapy was reported to delay the development of overt bone metastases. Ongoing studies will provide further insight regarding the anticancer potential of bisphosphonates and other antiresorptive agents.
Collapse
Affiliation(s)
- Michael Gnant
- Department of Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Währinger Gürtel 18-20, A-1090 Vienna, Austria.
| | | |
Collapse
|
40
|
Combination therapy inhibits development and progression of mammary tumours in immunocompetent mice. Breast Cancer Res Treat 2011; 133:523-36. [DOI: 10.1007/s10549-011-1782-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 09/13/2011] [Indexed: 10/17/2022]
|
41
|
|
42
|
Hamilton E, Clay TM, Blackwell KL. New Perspectives on Zoledronic Acid in Breast Cancer: Potential Augmentation of Anticancer Immune Response. Cancer Invest 2011; 29:533-41. [DOI: 10.3109/07357907.2011.605413] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
43
|
Ressler S, Mlineritsch B, Greil R. Zoledronic acid for adjuvant use in patients with breast cancer. Expert Rev Anticancer Ther 2011; 11:333-49. [PMID: 21417849 DOI: 10.1586/era.11.13] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Zoledronic acid, a nitrogen-containing bisphosphonate, is firmly established in the management of metastatic bone disease. It inhibits farnesyl diphosphonate synthase within the mevalonate pathway and, through this mechanism, is a potent inhibitor of osteoclast-mediated bone resorption. In addition, there are preclinical data suggesting that farnesyl diphosphonate synthase inhibition by zoledronic acid has anti-tumor effects in breast cancer. Adjuvant therapies for early breast cancer are associated with substantial decreases in bone mineral density. Results from three clinical trials, ABCSG-12, Z-FAST and ZO-FAST, indicate that the addition of twice-yearly zoledronic acid to standard adjuvant endocrine therapy in premenopausal and postmenopausal patients with hormone receptor-positive breast cancer prevents cancer treatment-induced bone loss. Moreover, it is becoming evident that it may also exert anticancer effects in an estrogen-deprived state in the adjuvant and neoadjuvant setting. However, long-term side effects need to be taken into consideration for treatment decisions.
Collapse
Affiliation(s)
- Sigrun Ressler
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectious Diseases, Laboratory of Immunological and Molecular Cancer Research, Private Medical University Hospital, Salzburg, Austria
| | | | | |
Collapse
|
44
|
Abstract
Bone metastases result in considerable morbidity, often affecting quality of life and independence over years, and may place complex demands on health care resources. The bisphosphonates have been shown to reduce skeletal morbidity in multiple myeloma and solid tumours affecting bone by 30-50%. Quite appropriately, these agents are increasingly used alongside anticancer treatments to prevent skeletal complications and relieve bone pain. The use of bisphosphonates in early cancer has become increasingly important to prevent adverse effects of cancer treatments on bone health. These include chemotherapy induced ovarian failure and the use of aromatase inhibitors in breast cancer and androgen deprivation therapy in prostate cancer. Bisphosphonate strategies, similar to those used to treat post-menopausal osteoporosis, are the intervention of choice for patients with low bone mineral density or rapid bone loss, along with adequate calcium and vitamin D intake and a healthy lifestyle. There is a strong preclinical rationale for bisphosphonates to prevent metastasis, primarily through inhibition of the vicious cycle of metastasis within the microenvironment. Recent data suggest that adjuvant bisphosphonates, at least in some patient subgroups, may modify the course of the disease and disrupt the metastatic process, reducing the risks of disease recurrence. In comparison to most other cancer treatments, adverse events related to bisphosphonate therapy are generally mild and infrequent; thus, the benefits of treatment within licensed indications will almost always outweigh the risks.
Collapse
Affiliation(s)
- Robert E Coleman
- Academic Unit of Clinical Oncology, Weston Park Hospital, Sheffield, UK.
| | | |
Collapse
|
45
|
Aft R, Perez JR, Raje N, Hirsh V, Saad F. Could targeting bone delay cancer progression? Potential mechanisms of action of bisphosphonates. Crit Rev Oncol Hematol 2011; 82:233-48. [PMID: 21683613 DOI: 10.1016/j.critrevonc.2011.05.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 05/18/2011] [Accepted: 05/25/2011] [Indexed: 12/12/2022] Open
Abstract
Although dissemination may occur early in the course of many cancers, the development of overt metastases depends upon a variety of factors inherent to the cancer cells and the tissue(s) they colonize. The time lag between initial dissemination and established metastases could be several years, during which period the bone marrow may provide an unwitting sanctuary for disseminated tumor cells (DTCs). Survival in a dormant state within the bone marrow may help DTCs weather the effects of anticancer therapies and seed posttreatment relapses. The importance of the bone marrow for facilitating DTC survival may vary depending on the type of cancer and mechanisms of tumor cell dissemination. By altering the bone microenvironment, bisphosphonates may reduce DTC viability. Moreover, some bisphosphonates have demonstrated multiple anticancer activities. These multiple mechanisms may help explain the improvement in disease outcomes with the use of zoledronic acid in malignancies like breast cancer and multiple myeloma.
Collapse
Affiliation(s)
- Rebecca Aft
- Department of Surgery, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| | | | | | | | | |
Collapse
|
46
|
Hafeman SD, Varland D, Dow SW. Bisphosphonates significantly increase the activity of doxorubicin or vincristine against canine malignant histiocytosis cells. Vet Comp Oncol 2011; 10:44-56. [PMID: 22236140 DOI: 10.1111/j.1476-5829.2011.00274.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Canine malignant histiocytosis (MH) is an aggressive neoplasm of macrophages and dendritic cells. It carries a poor prognosis because of the development of widespread metastasis and poor sensitivity to chemotherapy. Thus, there is a large need for new treatments for MH. We hypothesized that bisphosphonates might be useful to increase the effectiveness of cytotoxic chemotherapy against MH. To address this question, we conducted in vitro screening studies using MH cell lines and a panel of 6 chemotherapy and 5 bisphosphonate drugs. The combination of clodronate with vincristine was found to elicit synergistic killing which was associated with a significant increase in cell cycle arrest. Second, zoledronate combined with doxorubicin also significantly increased cell killing. Zoledronate significantly increased the uptake of doxorubicin by MH cells. On the basis of these findings, we conclude that certain bisphosphonate drugs may increase the overall effectiveness of chemotherapy for MH in dogs.
Collapse
Affiliation(s)
- S D Hafeman
- Department of Clinical Sciences, Animal Cancer Center, Colorado State University, Fort Collins, CO 80523, USA
| | | | | |
Collapse
|
47
|
De Luca A, Lamura L, Gallo M, Daniele G, D'Alessio A, Giordano P, Maiello MR, Pergameno M, Perrone F, Normanno N. Pharmacokinetic evaluation of zoledronic acid. Expert Opin Drug Metab Toxicol 2011; 7:911-8. [DOI: 10.1517/17425255.2011.585156] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
48
|
Mahtani R, Khan R, Jahanzeb M. The potential application of zoledronic acid as anticancer therapy in patients with non-small-cell lung cancer. Clin Lung Cancer 2011; 12:26-32. [PMID: 21273176 DOI: 10.3816/clc.2011.n.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Non-small-cell lung cancer (NSCLC) is frequently characterized by metastases to bone. Bisphosphonates have demonstrated efficacy in reducing the risk of skeletal-related events in cancer patients with bone metastases, including those with NSCLC. Zoledronic acid (ZA) is one of the most potent bisphosphonates and is approved for the first-line treatment of patients with multiple myeloma and bone metastases from solid tumors. Recent preclinical and clinical data suggest that ZA may also have direct and indirect anticancer effects. Several preclinical studies have provided insight into the potential mechanisms responsible for the anticancer activity of ZA, including inhibiting farnesyl pyrophosphate or geranylgeranyl pyrophosphate and activation of immune-mediated anticancer response by γδ T cells. In patients with NSCLC, ZA has been shown to reduce vascular endothelial growth factor levels with a direct correlation to clinical response. Clinical studies in this setting have shown that ZA may also provide a survival benefit and prolong time to progression. Ongoing studies are evaluating the efficacy of ZA for anticancer activity and prevention of bone metastases. Bisphosphonates, particularly ZA, are generally well tolerated and may likely offer an adjunct therapeutic option for patients with NSCLC.
Collapse
Affiliation(s)
- Reshma Mahtani
- Division of Hematology/Oncology, Sylvester Comprehensive Cancer Center, University of Miami Health Systems, Deerfield Beach, FL 33442, USA.
| | | | | |
Collapse
|
49
|
Bosch-Barrera J, Merajver SD, Menéndez JA, Van Poznak C. Direct antitumour activity of zoledronic acid: preclinical and clinical data. Clin Transl Oncol 2011; 13:148-55. [DOI: 10.1007/s12094-011-0634-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
50
|
Lipton A. Bones, breasts, and bisphosphonates: rationale for the use of zoledronic acid in advanced and early breast cancer. BREAST CANCER-TARGETS AND THERAPY 2011; 3:1-7. [PMID: 24367171 DOI: 10.2147/bctt.s16774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Bisphosphonates inhibit osteoclast-mediated bone resorption, thereby inhibiting the release of growth factors necessary to promote cancer cell growth, differentiation, and tumor formation in bone. These agents have demonstrated efficacy for delaying the onset and reducing the incidence of skeletal-related events in the advanced breast cancer setting, and have been shown to prevent cancer therapy-induced bone loss in the early breast cancer setting. Emerging clinical data indicate that the role of bisphosphonates in advanced and early breast cancer is evolving. Retrospective analyses and recent clinical trial data show that zoledronic acid may improve outcomes in some patients with breast cancer. Data from ABCSG-12 and ZO-FAST suggest that zoledronic acid may improve disease-free survival in the adjuvant breast cancer setting in postmenopausal women or women with endocrine therapy-induced menopause, and recent data from a predefined subset of the AZURE trial added to the anticancer story. However, the overall negative AZURE trial also raises questions about the role of bisphosphonates as an anticancer agent in patients with breast cancer. Overall, these data suggest that the addition of zoledronic acid to established anticancer regimens may have potential anticancer benefits in specific patient populations, although more studies are required to define its role.
Collapse
Affiliation(s)
- Allan Lipton
- Milton S. Hershey Medical Center, Pennsylvania State University, Hershey, PA, USA
| |
Collapse
|