1
|
Nakamura M, Magara T, Yoshimitsu M, Kano S, Kato H, Yokota K, Okuda K, Morita A. Blockade of glucose-6-phosphate dehydrogenase induces immunogenic cell death and accelerates immunotherapy. J Immunother Cancer 2024; 12:e008441. [PMID: 39089738 PMCID: PMC11293396 DOI: 10.1136/jitc-2023-008441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/07/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Enhanced glucose metabolism has been reported in many cancers. Glucose-6-phosphate dehydrogenase (G6PD) is a rate-limiting enzyme involved in the pentose phosphate pathway, which maintains NADPH levels and protects cells from oxidative damage. We recently found that low G6PD expression correlates with active tumor immunity. However, the mechanism involving G6PD and tumor immunity remained unclear. METHODS We conducted in vitro studies using G6PD-knocked down malignant melanoma cells, pathway analysis using the GEO dataset, in vivo studies in combination with immune checkpoint inhibitors (ICIs) using a mouse melanoma model, and prognostic analysis in 42 melanoma patients and 30 lung cancer patients who were treated with ICIs. RESULTS Inhibition of G6PD, both chemically and genetically, has been shown to decrease the production of NADPH and reduce their oxidative stress tolerance. This leads to cell death, which is accompanied by the release of high mobility group box 1 and the translocation of calreticulin to the plasma membrane. These findings suggested that inhibiting G6PD can induce immunogenic cell death. In experiments with C57BL/6 mice transplanted with G6PD-knockdown B16 melanoma cells and treated with anti-PD-L1 antibody, a significant reduction in tumor size was observed. Interestingly, inhibiting G6PD in only a part of the lesions increased the sensitivity of other lesions to ICI. Additionally, out of 42 melanoma patients and 30 lung cancer patients treated with ICIs, those with low G6PD expression had a better prognosis than those with high G6PD expression (p=0.0473; melanoma, p=0.0287; lung cancer). CONCLUSION G6PD inhibition is a potent therapeutic strategy that triggers immunogenic cell death in tumors, significantly augmenting the efficacy of immunotherapies.
Collapse
Affiliation(s)
- Motoki Nakamura
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences and Medical School, Nagoya, Japan
| | - Tetsuya Magara
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences and Medical School, Nagoya, Japan
| | - Maki Yoshimitsu
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences and Medical School, Nagoya, Japan
| | - Shinji Kano
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences and Medical School, Nagoya, Japan
| | - Hiroshi Kato
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences and Medical School, Nagoya, Japan
| | - Keisuke Yokota
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Katsuhiro Okuda
- Department of Oncology, Immunology and Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Akimichi Morita
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences and Medical School, Nagoya, Japan
| |
Collapse
|
2
|
Alotaibi F, Alshammari K, Alotaibi BA, Alsaab H. Destabilizing the genome as a therapeutic strategy to enhance response to immune checkpoint blockade: a systematic review of clinical trials evidence from solid and hematological tumors. Front Pharmacol 2024; 14:1280591. [PMID: 38264532 PMCID: PMC10803447 DOI: 10.3389/fphar.2023.1280591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 12/11/2023] [Indexed: 01/25/2024] Open
Abstract
Background: Genomic instability is increased alterations in the genome during cell division and is common among most cancer cells. Genome instability enhances the risk of initial carcinogenic transformation, generating new clones of tumor cells, and increases tumor heterogeneity. Although genome instability contributes to malignancy, it is also an "Achilles' heel" that constitutes a therapeutically-exploitable weakness-when sufficiently advanced, it can intrinsically reduce tumor cell survival by creating DNA damage and mutation events that overwhelm the capacity of cancer cells to repair those lesions. Furthermore, it can contribute to extrinsic survival-reducing events by generating mutations that encode new immunogenic antigens capable of being recognized by the immune system, particularly when anti-tumor immunity is boosted by immunotherapy drugs. Here, we describe how genome-destabilization can induce immune activation in cancer patients and systematically review the induction of genome instability exploited clinically, in combination with immune checkpoint blockade. Methods: We performed a systematic review of clinical trials that exploited the combination approach to successfully treat cancers patients. We systematically searched PubMed, Cochrane Central Register of Controlled Trials, Clinicaltrials.gov, and publication from the reference list of related articles. The most relevant inclusion criteria were peer-reviewed clinical trials published in English. Results: We identified 1,490 studies, among those 164 were clinical trials. A total of 37 clinical trials satisfied the inclusion criteria and were included in the study. The main outcome measurements were overall survival and progression-free survival. The majority of the clinical trials (30 out of 37) showed a significant improvement in patient outcome. Conclusion: The majority of the included clinical trials reported the efficacy of the concept of targeting DNA repair pathway, in combination with immune checkpoint inhibitors, to create a "ring of synergy" to treat cancer with rational combinations.
Collapse
Affiliation(s)
- Faizah Alotaibi
- College of Science and Health Professions, King Saud Bin Abdulaziz University for Health Sciences, Alahsa, Saudi Arabia
- King Abdullah International Medical Research Center, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
| | - Kanaan Alshammari
- King Abdullah International Medical Research Center, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
- Oncology Department, King Abdulaziz Medical City, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Badi A. Alotaibi
- King Abdullah International Medical Research Center, Ministry of National Guard-Health Affairs, Riyadh, Saudi Arabia
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Hashem Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, Taif, Saudi Arabia
| |
Collapse
|
3
|
Zheng Y, Wang PP, Fu Y, Chen YY, Ding ZY. Zoledronic acid enhances the efficacy of immunotherapy in non-small cell lung cancer. Int Immunopharmacol 2022; 110:109030. [DOI: 10.1016/j.intimp.2022.109030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/01/2022] [Accepted: 07/02/2022] [Indexed: 12/09/2022]
|
4
|
The mevalonate pathway in breast cancer biology. Cancer Lett 2022; 542:215761. [DOI: 10.1016/j.canlet.2022.215761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 02/07/2023]
|
5
|
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
|
6
|
Cholesterol and beyond - The role of the mevalonate pathway in cancer biology. Biochim Biophys Acta Rev Cancer 2020; 1873:188351. [PMID: 32007596 DOI: 10.1016/j.bbcan.2020.188351] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/14/2020] [Accepted: 01/30/2020] [Indexed: 02/07/2023]
Abstract
Cancer is a multifaceted global disease. Transformation of a normal to a malignant cell takes several steps, including somatic mutations, epigenetic alterations, metabolic reprogramming and loss of cell growth control. Recently, the mevalonate pathway has emerged as a crucial regulator of tumor biology and a potential therapeutic target. This pathway controls cholesterol production and posttranslational modifications of Rho-GTPases, both of which are linked to several key steps of tumor progression. Inhibitors of the mevalonate pathway induce pleiotropic antitumor-effects in several human malignancies, identifying the pathway as an attractive candidate for novel therapies. In this review, we will provide an overview about the role and regulation of the mevalonate pathway in certain aspects of cancer initiation and progression and its potential for therapeutic intervention in oncology.
Collapse
|
7
|
La-Beck NM, Liu X, Shmeeda H, Shudde C, Gabizon AA. Repurposing amino-bisphosphonates by liposome formulation for a new role in cancer treatment. Semin Cancer Biol 2019; 68:175-185. [PMID: 31874280 DOI: 10.1016/j.semcancer.2019.12.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 12/15/2019] [Indexed: 12/12/2022]
Abstract
Amino-bisphosphonates (N-BPs) have been commercially available for over four decades and are used for the treatment of osteoporosis, Paget's disease, hypercalcemia of malignancy, and bone metastases derived from various cancer types. Zoledronate and alendronate, two of the most potent N-BPs, have demonstrated direct tumoricidal activity on tumor cells and immune modulatory effects on myeloid cells and T cells in vitro and in animal models of cancer. However, the rapid renal clearance and sequestration in mineral bone of these drugs in free form severely limit their systemic exposure and applications in cancer patients. Reformulation of N-BPs by encapsulation in liposomal nanoparticles addresses these pharmacokinetic barriers, and liposomal zoledronate and alendronate formulations have been found to increase the anticancer efficacy of cytotoxic chemotherapies and adoptive T cell immunotherapies in murine cancer models. Herein, we review the differences in pharmacology between N-BPs versus non-N-BPs (e.g., clodronate), free versus liposomal N-BP formulations, and targeted versus non-targeted liposomal N-BPs, and the clinical and preclinical evidence supporting a role for liposomal N-BPs in the treatment of cancer. We propose that pegylated liposomal alendronate (PLA) has the most potential for clinical translation based on favorable therapeutic index, ability to passively target and accumulate in tumors, proven biocompatibility of the liposome carrier, and preclinical anticancer efficacy.
Collapse
Affiliation(s)
- Ninh M La-Beck
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, 1718 Pine St, Abilene, TX 79601, USA; Department of Pharmacy Practice, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, 1718 Pin. St, Abilene, TX 79601, USA.
| | - Xinli Liu
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, College of Pharmacy, 4849 Calhoun Road, Houston, TX 77204, USA
| | - Hilary Shmeeda
- Center of Nano-oncology, Shaare Zedek Medical Center, Jerusalem, Israel.
| | - Claire Shudde
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Jerry H. Hodge School of Pharmacy, 1718 Pine St, Abilene, TX 79601, USA.
| | - Alberto A Gabizon
- Center of Nano-oncology, Shaare Zedek Medical Center and Hebrew University-School of Medicine, POB 3235, Jerusalem, 91031, Israel.
| |
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
|
González-Fernández Y, Brown HK, Patiño-García A, Heymann D, Blanco-Prieto MJ. Oral administration of edelfosine encapsulated lipid nanoparticles causes regression of lung metastases in pre-clinical models of osteosarcoma. Cancer Lett 2018; 430:193-200. [PMID: 29802930 DOI: 10.1016/j.canlet.2018.05.030] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 05/17/2018] [Accepted: 05/18/2018] [Indexed: 12/14/2022]
Abstract
Osteosarcoma (OS) is the most frequent paediatric bone cancer, responsible for 9% of all cancer-related deaths in children. In this paper, a new strategy based on delivering edelfosine (ET) in lipid nanoparticles (LN) was explored in order to target the primary tumour and eliminate metastases. The in vitro and in vivo efficacy of the free drug, drug loaded into lipid nanoparticles (ET-LN) and doxorubicin (DOX) against osteosarcoma (OS) cells was analysed. ET and ET-LN decreased the growth of OS cells in vitro in a time- and dose-dependent manner. Interestingly, the uptake of ET and ET-LN was lower when OS cells were pre-treated with DOX. In vivo studies revealed that ET and ET-LN slowed down the primary tumour growth in two OS models. However, the combination of both drugs showed no additional anti-tumour effect. Importantly, ET-LN successfully prevented the metastatic spread of OS cells from the primary tumour to the lungs. On the whole, ET-LN are a promising candidate for OS chemotherapy.
Collapse
Affiliation(s)
- Yolanda González-Fernández
- Department of Pharmacy and Pharmaceutical Technology, University of Navarra, Irunlarrea 1, 31008, Pamplona, Spain; Laboratory of Pediatrics, University Clinic of Navarra, 31008, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, Irunlarrea 3, Pamplona, Spain
| | - Hannah K Brown
- INSERM, European Associated Laboratory "Sarcoma Research Unit", Department of Oncology and Metabolism, Medical School, University of Sheffield, UK
| | - Ana Patiño-García
- Laboratory of Pediatrics, University Clinic of Navarra, 31008, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, Irunlarrea 3, Pamplona, Spain
| | - Dominique Heymann
- INSERM, European Associated Laboratory "Sarcoma Research Unit", Department of Oncology and Metabolism, Medical School, University of Sheffield, UK; Institut de Cancérologie de l'Ouest, INSERM, U1232, CRCINA, Université de Nantes, Université d'Angers, 44805, cedex, Saint Herblain, France.
| | - María J Blanco-Prieto
- Department of Pharmacy and Pharmaceutical Technology, University of Navarra, Irunlarrea 1, 31008, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra, IdiSNA, Irunlarrea 3, Pamplona, Spain.
| |
Collapse
|
10
|
Rumney RMH, Coffelt SB, Neale TA, Dhayade S, Tozer GM, Miller G. PyMT-Maclow: A novel, inducible, murine model for determining the role of CD68 positive cells in breast tumor development. PLoS One 2017; 12:e0188591. [PMID: 29220404 PMCID: PMC5722323 DOI: 10.1371/journal.pone.0188591] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 11/09/2017] [Indexed: 02/02/2023] Open
Abstract
CD68+ tumor-associated macrophages (TAMs) are pro-tumorigenic, pro-angiogenic and are associated with decreased survival rates in patients with cancer, including breast cancer. Non-specific models of macrophage ablation reduce the number of TAMs and limit the development of mammary tumors. However, the lack of specificity and side effects associated with these models compromise their reliability. We hypothesized that specific and controlled macrophage depletion would provide precise data on the effects of reducing TAM numbers on tumor development. In this study, the MacLow mouse model of doxycycline-inducible and selective CD68+ macrophage depletion was crossed with the murine mammary tumor virus (MMTV)-Polyoma virus middle T antigen (PyMT) mouse model of spontaneous ductal breast adenocarcinoma to generate the PyMT-MacLow line. In doxycycline-treated PyMT-MacLow mice, macrophage numbers were decreased in areas surrounding tumors by 43%. Reducing the number of macrophages by this level delayed tumor progression, generated less proliferative tumors, decreased the vascularization of carcinomas and down-regulated the expression of many pro-angiogenic genes. These results demonstrate that depleting CD68+ macrophages in an inducible and selective manner delays the development of mammary tumors and that the PyMT-MacLow model is a useful and unique tool for studying the role of TAMs in breast cancer.
Collapse
Affiliation(s)
- Robin M. H. Rumney
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Seth B. Coffelt
- Department of Infection & Immunity, University of Sheffield, Sheffield, United Kingdom
- University of Glasgow, Institute of Cancer Sciences, Glasgow, United Kingdom
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
- * E-mail: (GM); (SC)
| | - Terence A. Neale
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Sandeep Dhayade
- Cancer Research UK Beatson Institute, Glasgow, United Kingdom
| | - Gillian M. Tozer
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
| | - Gaynor Miller
- Department of Oncology and Metabolism, University of Sheffield, Sheffield, United Kingdom
- * E-mail: (GM); (SC)
| |
Collapse
|
11
|
Shmeeda H, Amitay Y, Gorin J, Tzemach D, Mak L, Stern ST, Barenholz Y, Gabizon A. Coencapsulation of alendronate and doxorubicin in pegylated liposomes: a novel formulation for chemoimmunotherapy of cancer. J Drug Target 2016; 24:878-889. [PMID: 27187807 DOI: 10.1080/1061186x.2016.1191081] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
We developed a pegylated liposome formulation of a dissociable salt of a nitrogen-containing bisphosphonate, alendronate (Ald), coencapsulated with the anthracycline, doxorubicin (Dox), a commonly used chemotherapeutic agent. Liposome-encapsulated ammonium Ald generates a gradient driving Dox into liposomes, forming a salt that holds both drugs in the liposome water phase. The resulting formulation (PLAD) allows for a high-loading efficiency of Dox, comparable to that of clinically approved pegylated liposomal doxorubicin sulfate (PLD) and is very stable in plasma stability assays. Cytotoxicity tests indicate greater potency for PLAD compared to PLD. This appears to be related to a synergistic effect of the coencapsulated Ald and Dox. PLAD and PLD differed in in vitro monocyte-induced IL-1β release (greater for PLAD) and complement activation (greater for PLD). A molar ratio Ald/Dox of ∼1:1 seems to provide an optimal compromise between loading efficiency of Dox, circulation time and in vivo toxicity of PLAD. In mice, the circulation half-life and tumor uptake of PLAD were comparable to PLD. In the M109R and 4T1 tumor models in immunocompetent mice, PLAD was superior to PLD in the growth inhibition of subcutaneous tumor implants. This new formulation appears to be a promising tool to exploit the antitumor effects of aminobisphosphonates in synergy with chemotherapy.
Collapse
Affiliation(s)
| | - Yasmine Amitay
- a Shaare Zedek Medical Center , Jerusalem , Israel.,b School of Medicine, Hebrew University , Jerusalem , Israel
| | - Jenny Gorin
- a Shaare Zedek Medical Center , Jerusalem , Israel
| | - Dina Tzemach
- a Shaare Zedek Medical Center , Jerusalem , Israel
| | - Lidia Mak
- a Shaare Zedek Medical Center , Jerusalem , Israel
| | - Stephan T Stern
- c Frederick National Laboratory for Cancer Research, NCL , Frederick , MD , USA
| | | | - Alberto Gabizon
- a Shaare Zedek Medical Center , Jerusalem , Israel.,b School of Medicine, Hebrew University , Jerusalem , Israel
| |
Collapse
|
12
|
Göbel A, Thiele S, Browne AJ, Rauner M, Zinna VM, Hofbauer LC, Rachner TD. Combined inhibition of the mevalonate pathway with statins and zoledronic acid potentiates their anti-tumor effects in human breast cancer cells. Cancer Lett 2016; 375:162-171. [DOI: 10.1016/j.canlet.2016.03.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/02/2016] [Accepted: 03/02/2016] [Indexed: 12/22/2022]
|
13
|
Abstract
In the context of breast cancer, the importance of the skeleton in the regulation of primary tumour development and as a site for subsequent metastasis is well characterized. Our understanding of the contributions made by the host bone and bone marrow cells increasingly demonstrates the extent of the interaction between tumour cells and normal host cells. As a result, the need to develop and utilize therapies that can impede the growth and/or function of tumour cells while sparing normal host bone and bone marrow cells is immense and expanding. The need for these new treatments is, however, superimposed on the orthopaedic management of patients' quality of life, where pain control and continued locomotion are paramount. Indeed, the majority of the anticancer therapies used to date often result in direct or indirect damage to bone. Thus, although the bone microenvironment regulates tumour cell growth in bone, cells within the bone marrow niche also mediate many of the orthopaedic consequences of tumour progression as well as resistance to the antitumour effects of existing therapies. In this Review, we highlight the effects of existing cancer treatments on bone and the bone marrow microenvironment as well as the mechanisms mediating these effects and the current utility of modern orthopaedic interventions.
Collapse
Affiliation(s)
- Issam Makhoul
- Department of Medicine, Division of Haematology/Oncology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA
| | - Corey O Montgomery
- Department of Orthopaedic Surgery, Centre for Orthopaedic Research, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA
| | - Dana Gaddy
- Department of Physiology and Biophysics, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA
| | - Larry J Suva
- Department of Orthopaedic Surgery, Centre for Orthopaedic Research, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, AR 72205, USA
| |
Collapse
|
14
|
Kroep JR, Charehbili A, Coleman RE, Aft RL, Hasegawa Y, Winter MC, Weilbaecher K, Akazawa K, Hinsley S, Putter H, Liefers GJ, Nortier JWR, Kohno N. Effects of neoadjuvant chemotherapy with or without zoledronic acid on pathological response: A meta-analysis of randomised trials. Eur J Cancer 2015; 54:57-63. [PMID: 26722766 PMCID: PMC4928630 DOI: 10.1016/j.ejca.2015.10.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2015] [Revised: 10/12/2015] [Accepted: 10/17/2015] [Indexed: 11/30/2022]
Abstract
Purpose The addition of bisphosphonates to adjuvant therapy improves survival in postmenopausal breast cancer (BC) patients. We report a meta-analysis of four randomised trials of neoadjuvant chemotherapy (CT) +/− zoledronic acid (ZA) in stage II/III BC to investigate the potential for enhancing the pathological response. Methods Individual patient data from four prospective randomised clinical trials reporting the effect of the addition of ZA on the pathological response after neoadjuvant CT were pooled. Primary outcomes were pathological complete response in the breast (pCRb) and in the breast and lymph nodes (pCR). Trial-level and individual patient data meta-analyses were done. Predefined subgroup-analyses were performed for postmenopausal women and patients with triple-negative BC. Results pCRb and pCR data were available in 735 and 552 patients respectively. In the total study population ZA addition to neoadjuvant CT did not increase pCRb or pCR rates. However, in postmenopausal patients, the addition of ZA resulted in a significant, near doubling of the pCRb rate (10.8% for CT only versus 17.7% with CT+ZA; odds ratio [OR] 2.14, 95% confidence interval [CI] 1.01–4.55) and a non-significant benefit of the pCR rate (7.8% for CT only versus 14.6% with CT+ZA; OR 2.62, 95% CI 0.90–7.62). In patients with triple-negative BC a trend was observed favouring CT+ZA. Conclusion This meta-analysis shows no impact from the addition of ZA to neoadjuvant CT on pCR. However, as has been seen in the adjuvant setting, the addition of ZA to neoadjuvant CT may augment the effects of CT in postmenopausal patients with BC.
Collapse
Affiliation(s)
- J R Kroep
- Leiden University Medical Center, Leiden, The Netherlands
| | - A Charehbili
- Leiden University Medical Center, Leiden, The Netherlands
| | - R E Coleman
- Sheffield Cancer Research Centre, Weston Park Hospital, Sheffield, United Kingdom
| | - R L Aft
- Washington University School of Medicine, St. Louis, USA
| | - Y Hasegawa
- Hirosaki Municipal Hospital, Aomori, Japan
| | - M C Winter
- Sheffield Cancer Research Centre, Weston Park Hospital, Sheffield, United Kingdom
| | - K Weilbaecher
- Washington University School of Medicine, St. Louis, USA
| | | | - S Hinsley
- Clinical Trials Research Unit, Leeds, United Kingdom
| | - H Putter
- Leiden University Medical Center, Leiden, The Netherlands
| | - G J Liefers
- Leiden University Medical Center, Leiden, The Netherlands
| | - J W R Nortier
- Leiden University Medical Center, Leiden, The Netherlands
| | - N Kohno
- Kobe Kaisei Hospital, Kobe, Japan
| |
Collapse
|
15
|
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
|
16
|
Vrdoljak E, Gore M, Leyman S, Szczylik C, Kharkevich G, Schöffski P, Torday L, Mardiak J, Zhang K, Sajben P, Sella A. Bisphosphonates in patients with renal cell carcinoma and bone metastases: a sunitinib global expanded-access trial subanalysis. Future Oncol 2015; 11:2831-40. [PMID: 26118456 DOI: 10.2217/fon.15.140] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To investigate retrospectively the effects of bone metastases and bisphosphonates in sunitinib-treated metastatic renal cell carcinoma patients. PATIENTS & METHODS Patients in Groups (Gp) 1 and 2, but not Gp3, had bone metastases. Gp2 received bisphosphonates following standard practice. RESULTS Gp2 had less favorable prognosis than Gp1. Gp3 had fewer metastases and the best prognosis. More serious adverse events occurred in Gp2 versus Gp1. The difference in overall survival between Gp1 and Gp2 was not significant after adjusting for covariates. Significantly shorter overall survival in Gp1 versus Gp3 persisted after adjusting for covariates. CONCLUSION Bone metastases may have a negative prognostic impact in metastatic renal cell carcinoma. Bisphosphonates may have delayed early disease progression for prognostically worse sunitinib/bisphosphonate-treated patients.
Collapse
Affiliation(s)
- Eduard Vrdoljak
- Centre of Oncology, Medical School Split, University Hospital Split, Split, Spinciceva 1, 21000 Split, Croatia
| | - Martin Gore
- Royal Marsden Hospital, Fulham Road, London, SW3 6JJ, UK
| | - Sophie Leyman
- Pfizer, Boulevard de la Plaine 17, 1050 Elsene/Ixelles (Brussels), Belgium
| | - Cezary Szczylik
- Department of Oncology, Central Clinical Hospital, Military Institute of Medicine, Szaserόw 128, 00-909 Warsaw, Poland
| | - Galina Kharkevich
- Biotherapy Department, N. N. Blokhin Russian Cancer Research Centre, 24 Kashirskoe Shosse, Moscow 115478, Russia
| | - Patrick Schöffski
- University Hospitals Leuven, Leuven Cancer Institute, Herestraat 49, 3000 Leuven, Belgium
| | - Laszlo Torday
- Department of Oncotherapy, University of Szeged, Korányi fasor 12, 6720, Szeged, Hungary
| | - Josef Mardiak
- Department of Medical Oncology, National Cancer Institute, Vlárska 7, 833 91 Bratislava, Slovakia
| | - Ke Zhang
- Pfizer, 10646 Science Center Drive, La Jolla, San Diego, CA 92121, USA
| | - Peter Sajben
- Pfizer, 235 East 42nd Street, New York, NY 10017, USA
| | - Avishay Sella
- Department of Oncology, Assaf Harofeh Medical Center Zerifin, Beer Yacov, Tel Aviv 70300, Israel
| |
Collapse
|
17
|
HATTORI YOSHIYUKI, SHIBUYA KAZUHIKO, KOJIMA KAORI, MIATMOKO ANDANG, KAWANO KUMI, OZAKI KEIICHI, YONEMOCHI ETSUO. Zoledronic acid enhances antitumor efficacy of liposomal doxorubicin. Int J Oncol 2015; 47:211-9. [DOI: 10.3892/ijo.2015.2991] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 04/02/2015] [Indexed: 11/05/2022] Open
|
18
|
Ottewell PD, Wang N, Meek J, Fowles CA, Croucher PI, Eaton CL, Holen I. Castration-induced bone loss triggers growth of disseminated prostate cancer cells in bone. Endocr Relat Cancer 2014; 21:769-81. [PMID: 25052474 DOI: 10.1530/erc-14-0199] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Up to 90% of patients with castrate-resistant prostate cancer develop bone metastases, and the majority of these men have received androgen deprivation therapy known to cause bone loss. Whether this treatment-induced change to the bone microenvironment affects disseminated tumour cells, potentially stimulating development of bone metastasis, remains to be determined. The objective of this study was to use an in vivo model mimicking androgen ablation to establish the effects of this intervention on disseminated prostate cancer cells in bone. We mimicked the effects of androgen deprivation on bone metastasis by castrating 12-week-old BALB/c nude mice that had disseminated, hormone-insensitive PC3 prostate cancer cells present in the long bones. Castration caused increased bone resorption and loss of bone volume, compared with sham operation. In addition, castration triggered growth of disseminated PC3 cells to form bone metastasis in 70% of animals. In contrast, only 10% of sham-operated animals had detectable long bone tumours. Weekly administration of 100 μg/kg zoledronic acid (ZOL) prevented castration-induced tumour growth in bone and increased bone volume, but did not eliminate the disseminated tumour cells. ZOL had no effect on tumour growth in the sham-operated animals, despite causing a significant increase in bone volume. This is the first demonstration that, in a model of prostate cancer bone metastasis, mimicking androgen ablation results in growth of disseminated tumour cells in bone through osteoclast-mediated mechanisms. We provide the first biological evidence supporting the administration of ZOL to prostate cancer patients at the time of androgen ablation to prevent subsequent relapse in bone.
Collapse
Affiliation(s)
- Penelope D Ottewell
- Academic Unit of Clinical Oncology, Department of OncologyBone Biology, Department of Human Metabolism, Medical School, University of SheffieldBeech Hill Road, Sheffield S10 2RX, UKMusculoskeletal Medicine DivisionGarvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Ning Wang
- Academic Unit of Clinical Oncology, Department of OncologyBone Biology, Department of Human Metabolism, Medical School, University of SheffieldBeech Hill Road, Sheffield S10 2RX, UKMusculoskeletal Medicine DivisionGarvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Joshua Meek
- Academic Unit of Clinical Oncology, Department of OncologyBone Biology, Department of Human Metabolism, Medical School, University of SheffieldBeech Hill Road, Sheffield S10 2RX, UKMusculoskeletal Medicine DivisionGarvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - C Anne Fowles
- Academic Unit of Clinical Oncology, Department of OncologyBone Biology, Department of Human Metabolism, Medical School, University of SheffieldBeech Hill Road, Sheffield S10 2RX, UKMusculoskeletal Medicine DivisionGarvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Peter I Croucher
- Academic Unit of Clinical Oncology, Department of OncologyBone Biology, Department of Human Metabolism, Medical School, University of SheffieldBeech Hill Road, Sheffield S10 2RX, UKMusculoskeletal Medicine DivisionGarvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Colby L Eaton
- Academic Unit of Clinical Oncology, Department of OncologyBone Biology, Department of Human Metabolism, Medical School, University of SheffieldBeech Hill Road, Sheffield S10 2RX, UKMusculoskeletal Medicine DivisionGarvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Ingunn Holen
- Academic Unit of Clinical Oncology, Department of OncologyBone Biology, Department of Human Metabolism, Medical School, University of SheffieldBeech Hill Road, Sheffield S10 2RX, UKMusculoskeletal Medicine DivisionGarvan Institute of Medical Research, Sydney, New South Wales, Australia
| |
Collapse
|
19
|
Levin-Gromiko U, Koshelev V, Kushnir P, Fedida-Metula S, Voronov E, Fishman D. Amplified lipid rafts of malignant cells constitute a target for inhibition of aberrantly active NFAT and melanoma tumor growth by the aminobisphosphonate zoledronic acid. Carcinogenesis 2014; 35:2555-66. [DOI: 10.1093/carcin/bgu178] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
20
|
Rogers TL, Wind N, Hughes R, Nutter F, Brown HK, Vasiliadou I, Ottewell PD, Holen I. Macrophages as potential targets for zoledronic acid outside the skeleton-evidence from in vitro and in vivo models. Cell Oncol (Dordr) 2013; 36:505-14. [PMID: 24177992 DOI: 10.1007/s13402-013-0156-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/11/2013] [Indexed: 12/19/2022] Open
Abstract
PURPOSE Multiple cell types of the tumour microenvironment, including macrophages, contribute to the response to cancer therapy. The anti-resorptive agent zoledronic acid (ZOL) has anti-tumour effects in vitro and in vivo, but it is not known to what extent macrophages are affected by this agent. We have therefore investigated the effects of ZOL on macrophages using a combination of in vitro and in vivo models. METHODS J774 macrophages were treated with ZOL in vitro, alone and in combination with doxorubicin (DOX), and the levels of apoptosis and necrosis determined. Uptake of zoledronic acid was assessed by detection of unprenylated Rap1a in J774 macrophages in vitro, in peritoneal macrophages and in macrophage populations isolated from subcutaneously implanted breast cancer xenografts following ZOL treatment in vivo. RESULTS Exposure of J774 macrophages to 5 μM ZOL for 24 h caused a significant increase in the levels of uRap1A, and higher doses/longer exposure induced apoptotic cell death. DOX (10 nM/24 h) and ZOL (10 μM/4 h) given in sequence induced significantly increased levels of apoptotic cell death compared to single agents. Peritoneal macrophages and macrophage populations isolated from breast tumour xenografts had detectable levels of uRap1A 24 h following a single, clinically achievable dose of 100 μg/kg ZOL in vivo. CONCLUSION We demonstrate that macrophages are sensitive to sequential administration of DOX and ZOL, and that both peritoneal and breast tumour associated macrophages rapidly take up ZOL in vivo. Our data support that macrophages may contribute to the anti-tumour effect of ZOL.
Collapse
Affiliation(s)
- T L Rogers
- Department of Oncology, CR-UK/YCR Sheffield Cancer Research Centre, University of Sheffield, Sheffield, UK
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Choi M, Shin DH, Kim JS. Repositioning of zoledronic acid for breast cancer using transferrin-conjugated liposome. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2013. [DOI: 10.1007/s40005-013-0091-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
22
|
Winter MC, Wilson C, Syddall SP, Cross SS, Evans A, Ingram CE, Jolley IJ, Hatton MQ, Freeman JV, Mori S, Holen I, Coleman RE. Neoadjuvant Chemotherapy with or without Zoledronic Acid in Early Breast Cancer—A Randomized Biomarker Pilot Study. Clin Cancer Res 2013; 19:2755-65. [DOI: 10.1158/1078-0432.ccr-12-3235] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
23
|
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
|
24
|
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
|