1
|
Adams A, Jakob T, Huth A, Monsef I, Ernst M, Kopp M, Caro-Valenzuela J, Wöckel A, Skoetz N. Bone-modifying agents for reducing bone loss in women with early and locally advanced breast cancer: a network meta-analysis. Cochrane Database Syst Rev 2024; 7:CD013451. [PMID: 38979716 PMCID: PMC11232105 DOI: 10.1002/14651858.cd013451.pub2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
BACKGROUND Bisphosphonates and receptor activator of nuclear factor-kappa B ligand (RANKL)-inhibitors are amongst the bone-modifying agents used as supportive treatment in women with breast cancer who do not have bone metastases. These agents aim to reduce bone loss and the risk of fractures. Bisphosphonates have demonstrated survival benefits, particularly in postmenopausal women. OBJECTIVES To assess and compare the effects of different bone-modifying agents as supportive treatment to reduce bone mineral density loss and osteoporotic fractures in women with breast cancer without bone metastases and generate a ranking of treatment options using network meta-analyses (NMAs). SEARCH METHODS We identified studies by electronically searching CENTRAL, MEDLINE and Embase until January 2023. We searched various trial registries and screened abstracts of conference proceedings and reference lists of identified trials. SELECTION CRITERIA We included randomised controlled trials comparing different bisphosphonates and RANKL-inihibitors with each other or against no further treatment or placebo for women with breast cancer without bone metastases. DATA COLLECTION AND ANALYSIS Two review authors independently extracted data and assessed the risk of bias of included studies and certainty of evidence using GRADE. Outcomes were bone mineral density, quality of life, overall fractures, overall survival and adverse events. We conducted NMAs and generated treatment rankings. MAIN RESULTS Forty-seven trials (35,163 participants) fulfilled our inclusion criteria; 34 trials (33,793 participants) could be considered in the NMA (8 different treatment options). Bone mineral density We estimated that the bone mineral density of participants with no treatment/placebo measured as total T-score was -1.34. Evidence from the NMA (9 trials; 1166 participants) suggests that treatment with ibandronate (T-score -0.77; MD 0.57, 95% CI -0.05 to 1.19) may slightly increase bone mineral density (low certainty) and treatment with zoledronic acid (T-score -0.45; MD 0.89, 95% CI 0.62 to 1.16) probably slightly increases bone mineral density compared to no treatment/placebo (moderate certainty). Risedronate (T-score -1.08; MD 0.26, 95% CI -0.32 to 0.84) may result in little to no difference compared to no treatment/placebo (low certainty). We are uncertain whether alendronate (T-score 2.36; MD 3.70, 95% CI -2.01 to 9.41) increases bone mineral density compared to no treatment/placebo (very low certainty). Quality of life No quantitative analyses could be performed for quality of life, as only three studies reported this outcome. All three studies showed only minimal differences between the respective interventions examined. Overall fracture rate We estimated that 70 of 1000 participants with no treatment/placebo had fractures. Evidence from the NMA (16 trials; 19,492 participants) indicates that treatment with clodronate or ibandronate (42 of 1000; RR 0.60, 95% CI 0.39 to 0.92; 40 of 1000; RR 0.57, 95% CI 0.38 to 0.86, respectively) decreases the number of fractures compared to no treatment/placebo (high certainty). Denosumab or zoledronic acid (51 of 1000; RR 0.73, 95% CI 0.52 to 1.01; 55 of 1000; RR 0.79, 95% CI 0.56 to 1.11, respectively) probably slightly decreases the number of fractures; and risedronate (39 of 1000; RR 0.56, 95% CI 0.15 to 2.16) probably decreases the number of fractures compared to no treatment/placebo (moderate certainty). Pamidronate (106 of 1000; RR 1.52, 95% CI 0.75 to 3.06) probably increases the number of fractures compared to no treatment/placebo (moderate certainty). Overall survival We estimated that 920 of 1000 participants with no treatment/placebo survived overall. Evidence from the NMA (17 trials; 30,991 participants) suggests that clodronate (924 of 1000; HR 0.95, 95% CI 0.77 to 1.17), denosumab (927 of 1000; HR 0.91, 95% CI 0.69 to 1.21), ibandronate (915 of 1000; HR 1.06, 95% CI 0.83 to 1.34) and zoledronic acid (925 of 1000; HR 0.93, 95% CI 0.76 to 1.14) may result in little to no difference regarding overall survival compared to no treatment/placebo (low certainty). Additionally, we are uncertain whether pamidronate (905 of 1000; HR 1.20, 95% CI 0.81 to 1.78) decreases overall survival compared to no treatment/placebo (very low certainty). Osteonecrosis of the jaw We estimated that 1 of 1000 participants with no treatment/placebo developed osteonecrosis of the jaw. Evidence from the NMA (12 trials; 23,527 participants) suggests that denosumab (25 of 1000; RR 24.70, 95% CI 9.56 to 63.83), ibandronate (6 of 1000; RR 5.77, 95% CI 2.04 to 16.35) and zoledronic acid (9 of 1000; RR 9.41, 95% CI 3.54 to 24.99) probably increases the occurrence of osteonecrosis of the jaw compared to no treatment/placebo (moderate certainty). Additionally, clodronate (3 of 1000; RR 2.65, 95% CI 0.83 to 8.50) may increase the occurrence of osteonecrosis of the jaw compared to no treatment/placebo (low certainty). Renal impairment We estimated that 14 of 1000 participants with no treatment/placebo developed renal impairment. Evidence from the NMA (12 trials; 22,469 participants) suggests that ibandronate (28 of 1000; RR 1.98, 95% CI 1.01 to 3.88) probably increases the occurrence of renal impairment compared to no treatment/placebo (moderate certainty). Zoledronic acid (21 of 1000; RR 1.49, 95% CI 0.87 to 2.58) probably increases the occurrence of renal impairment while clodronate (12 of 1000; RR 0.88, 95% CI 0.55 to 1.39) and denosumab (11 of 1000; RR 0.80, 95% CI 0.54 to 1.19) probably results in little to no difference regarding the occurrence of renal impairment compared to no treatment/placebo (moderate certainty). AUTHORS' CONCLUSIONS When considering bone-modifying agents for managing bone loss in women with early or locally advanced breast cancer, one has to balance between efficacy and safety. Our findings suggest that bisphosphonates (excluding alendronate and pamidronate) or denosumab compared to no treatment or placebo likely results in increased bone mineral density and reduced fracture rates. Our survival analysis that included pre and postmenopausal women showed little to no difference regarding overall survival. These treatments may lead to more adverse events. Therefore, forming an overall judgement of the best ranked bone-modifying agent is challenging. More head-to-head comparisons, especially comparing denosumab with any bisphosphonate, are needed to address gaps and validate the findings of this review.
Collapse
Affiliation(s)
- Anne Adams
- Institute of Medical Statistics and Computational Biology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Tina Jakob
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Alessandra Huth
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Ina Monsef
- Cochrane Haematology, Institute of Public Health, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Moritz Ernst
- Cochrane Haematology, Institute of Public Health, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Marco Kopp
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Julia Caro-Valenzuela
- Cochrane Haematology, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Achim Wöckel
- Department of Gynaecology and Obstetrics, University Hospital of Würzburg, Würzburg, Germany
| | - Nicole Skoetz
- Cochrane Haematology, Institute of Public Health, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
2
|
Panagiotakou A, Yavropoulou M, Nasiri-Ansari N, Makras P, Basdra EK, Papavassiliou AG, Kassi EN. Extra-skeletal effects of bisphosphonates. Metabolism 2020; 110:154264. [PMID: 32445641 DOI: 10.1016/j.metabol.2020.154264] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/20/2020] [Accepted: 05/20/2020] [Indexed: 12/21/2022]
Abstract
Bisphosphonates (BPs) are pyrophosphate analogues widely used in diseases related to bone loss and increased bone turnover. Their high affinity for bone hydroxyapatite makes them ideal agents for bone diseases, while preventing them from reaching other cells and tissues. Data of the last decade, however, have demonstrated extra-skeletal tissue deposition and a variety of non-skeletal effects have been recently recognized. As such, BPs have been shown to exert anti-tumor, immunomodulatory, anti-inflammatory and anti-diabetic effects. In addition, new delivery systems (liposomes, nanoparticles, hydrogels) are being developed in an effort to expand BPs clinical application to extra-skeletal tissues and enhance their overall therapeutic spectrum and effectiveness. In the present review, we outline current data on extra-skeletal actions of bisphosphonates and attempt to unravel the underlying pathophysiological mechanisms.
Collapse
Affiliation(s)
- Argyro Panagiotakou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Greece
| | - Maria Yavropoulou
- 1st Department of Propaedeutic Internal Medicine, Endocrinology Unit, "Laiko" General Hospital, Medical School, National and Kapodistrian University of Athens, Greece
| | - Narjes Nasiri-Ansari
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Greece.
| | - Polyzois Makras
- Department of Medical Research, 251 Hellenic Air Force General Hospital, Athens, Greece
| | - Efthimia K Basdra
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Greece.
| | - Eva N Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Greece; 1st Department of Propaedeutic Internal Medicine, Endocrinology Unit, "Laiko" General Hospital, Medical School, National and Kapodistrian University of Athens, Greece.
| |
Collapse
|
3
|
Comparative Study of Neoadjuvant Chemotherapy With and Without Zometa for Management of Locally Advanced Breast Cancer With Serum VEGF as Primary Endpoint: The NEOZOL Study. Clin Breast Cancer 2018; 18:e1311-e1321. [PMID: 30098917 DOI: 10.1016/j.clbc.2018.07.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 06/22/2018] [Accepted: 07/05/2018] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Neoadjuvant chemotherapy has become the treatment of choice for locally advanced breast cancer. Zoledronic acid (ZA) is a bisphosphonate initially used in the treatment of bone metastases because of its antibone resorption effect. Antitumor effects of ZA, including the inhibition of cell adhesion to mineralized bone or the antiangiogenic effect, have been demonstrated. However, the clinical significance of these effects remains to be determined. MATERIALS AND METHODS We undertook a multicenter open-label randomized trial to analyze the value of adding ZA to neoadjuvant chemotherapy for TNM clinical stage T2/T3 breast cancer. The primary endpoint was the evolution of serum VEGF. RESULTS The data from 24 patients were included in the ZA group and 26 in the control group. The evolution of serum VEGF was slightly in favor of ZA at 5.5 months (-0.7% vs. +7.5%), without reaching statistical significance (P = .52). The secondary endpoints were the breast conservation rate (higher with ZA; 83.3% vs. 65.4%; P = NS), pathologic complete response (no effect), and circulating tumor cells (odds ratio, 0.68 in favor of ZA; 95% confidence interval, 0.02-24.36). No cases of jaw necrosis or severe renal failure were observed in either group. CONCLUSION ZA is an antitumor drug of interest because of its multiple effects on tumor biology. Larger trials with longer follow-up that include additional endpoints such as relapse and survival rates would be of interest.
Collapse
|
4
|
Sousa S, Clézardin P. Bone-Targeted Therapies in Cancer-Induced Bone Disease. Calcif Tissue Int 2018; 102:227-250. [PMID: 29079995 DOI: 10.1007/s00223-017-0353-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 10/19/2017] [Indexed: 01/14/2023]
Abstract
Cancer-induced bone disease is a major source of morbidity and mortality in cancer patients. Thus, effective bone-targeted therapies are essential to improve disease-free, overall survival and quality of life of cancer patients with bone metastases. Depending of the cancer-type, bone metastases mainly involve the modulation of osteoclast and/or osteoblast activity by tumour cells. To inhibit metastatic bone disease effectively, it is imperative to understand its underlying mechanisms and identify the target cells for therapy. If the aim is to prevent bone metastasis, it is essential to target not only bone metastatic features in the tumour cells, but also tumour-nurturing bone microenvironment properties. The currently available bone-targeted agents mainly affect osteoclasts, inhibiting bone resorption (e.g. bisphosphonates, denosumab). Some agents targeting osteoblasts begin to emerge which target osteoblasts (e.g. romosozumab), activating bone formation. Moreover, certain drugs initially thought to target only osteoclasts are now known to have a dual action (activating osteoblasts and inhibiting osteoclasts, e.g. proteasome inhibitors). This review will focus on the evolution of bone-targeted therapies for the treatment of cancer-induced bone disease, summarizing preclinical and clinical findings obtained with anti-resorptive and bone anabolic therapies.
Collapse
Affiliation(s)
- Sofia Sousa
- National Institute of Health and Medical Research (INSERM), UMR 1033, 69372, Lyon, France.
- Faculty of Medicine Laennec, University of Lyon-1, 69372, Villeurbanne, France.
| | - Philippe Clézardin
- National Institute of Health and Medical Research (INSERM), UMR 1033, 69372, Lyon, France
- Faculty of Medicine Laennec, University of Lyon-1, 69372, Villeurbanne, France
- European Cancer and Bone Metastasis Laboratory, Department of Bone Oncology and Metabolism, Mellanby Centre for Bone Research, University of Sheffield, Sheffield, UK
| |
Collapse
|
5
|
O'Carrigan B, Wong MHF, Willson ML, Stockler MR, Pavlakis N, Goodwin A. Bisphosphonates and other bone agents for breast cancer. Cochrane Database Syst Rev 2017; 10:CD003474. [PMID: 29082518 PMCID: PMC6485886 DOI: 10.1002/14651858.cd003474.pub4] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Bone is the most common site of metastatic disease associated with breast cancer (BC). Bisphosphonates inhibit osteoclast-mediated bone resorption, and novel targeted therapies such as denosumab inhibit other key bone metabolism pathways. We have studied these agents in both early breast cancer and advanced breast cancer settings. This is an update of the review originally published in 2002 and subsequently updated in 2005 and 2012. OBJECTIVES To assess the effects of bisphosphonates and other bone agents in addition to anti-cancer treatment: (i) in women with early breast cancer (EBC); (ii) in women with advanced breast cancer without bone metastases (ABC); and (iii) in women with metastatic breast cancer and bone metastases (BCBM). SEARCH METHODS In this review update, we searched Cochrane Breast Cancer's Specialised Register, CENTRAL, MEDLINE, Embase, the World Health Organization's International Clinical Trials Registry Platform (WHO ICTRP) and ClinicalTrials.gov on 19 September 2016. SELECTION CRITERIA We included randomised controlled trials (RCTs) comparing: (a) one treatment with a bisphosphonate/bone-acting agent with the same treatment without a bisphosphonate/bone-acting agent; (b) treatment with one bisphosphonate versus treatment with a different bisphosphonate; (c) treatment with a bisphosphonate versus another bone-acting agent of a different mechanism of action (e.g. denosumab); and (d) immediate treatment with a bisphosphonate/bone-acting agent versus delayed treatment of the same bisphosphonate/bone-acting agent. DATA COLLECTION AND ANALYSIS Two review authors independently extracted data, and assessed risk of bias and quality of the evidence. The primary outcome measure was bone metastases for EBC and ABC, and a skeletal-related event (SRE) for BCBM. We derived risk ratios (RRs) for dichotomous outcomes and the meta-analyses used random-effects models. Secondary outcomes included overall survival and disease-free survival for EBC; we derived hazard ratios (HRs) for these time-to-event outcomes where possible. We collected toxicity and quality-of-life information. GRADE was used to assess the quality of evidence for the most important outcomes in each treatment setting. MAIN RESULTS We included 44 RCTs involving 37,302 women.In women with EBC, bisphosphonates were associated with a reduced risk of bone metastases compared to placebo/no bisphosphonate (RR 0.86, 95% confidence interval (CI) 0.75 to 0.99; P = 0.03, 11 studies; 15,005 women; moderate-quality evidence with no significant heterogeneity). Bisphosphonates provided an overall survival benefit with time-to-event data (HR 0.91, 95% CI 0.83 to 0.99; P = 0.04; 9 studies; 13,949 women; high-quality evidence with evidence of heterogeneity). Subgroup analysis by menopausal status showed a survival benefit from bisphosphonates in postmenopausal women (HR 0.77, 95% CI 0.66 to 0.90; P = 0.001; 4 studies; 6048 women; high-quality evidence with no evidence of heterogeneity) but no survival benefit for premenopausal women (HR 1.03, 95% CI 0.86 to 1.22; P = 0.78; 2 studies; 3501 women; high-quality evidence with no heterogeneity). There was evidence of no effect of bisphosphonates on disease-free survival (HR 0.94, 95% 0.87 to 1.02; P = 0.13; 7 studies; 12,578 women; high-quality evidence with significant heterogeneity present) however subgroup analyses showed a disease-free survival benefit from bisphosphonates in postmenopausal women only (HR 0.82, 95% CI 0.74 to 0.91; P < 0.001; 7 studies; 8314 women; high-quality evidence with no heterogeneity). Bisphosphonates did not significantly reduce the incidence of fractures when compared to placebo/no bisphosphonates (RR 0.77, 95% CI 0.54 to 1.08, P = 0.13, 6 studies, 7602 women; moderate-quality evidence due to wide confidence intervals). We await mature overall survival and disease-free survival results for denosumab trials.In women with ABC without clinically evident bone metastases, there was no evidence of an effect of bisphosphonates on bone metastases (RR 0.96, 95% CI 0.65 to 1.43; P = 0.86; 3 studies; 330 women; moderate-quality evidence with no heterogeneity) or overall survival (RR 0.89, 95% CI 0.73 to 1.09; P = 0.28; 3 studies; 330 women; high-quality evidence with no heterogeneity) compared to placebo/no bisphosphonates however the confidence intervals were wide. One study reported a trend towards an extended period of time without a SRE with bisphosphonate compared to placebo (low-quality evidence). One study reported quality of life and there was no apparent difference in scores between bisphosphonate and placebo (moderate-quality evidence).In women with BCBM, bisphosphonates reduced the SRE risk by 14% (RR 0.86, 95% CI 0.78 to 0.95; P = 0.003; 9 studies; 2810 women; high-quality evidence with evidence of heterogeneity) compared with placebo/no bisphosphonates. This benefit persisted when administering either intravenous or oral bisphosphonates versus placebo. Bisphosphonates delayed the median time to a SRE with a median ratio of 1.43 (95% CI 1.29 to 1.58; P < 0.00001; 9 studies; 2891 women; high-quality evidence with no heterogeneity) and reduced bone pain (in 6 out of 11 studies; moderate-quality evidence) compared to placebo/no bisphosphonate. Treatment with bisphosphonates did not appear to affect overall survival (RR 1.01, 95% CI 0.91 to 1.11; P = 0.85; 7 studies; 1935 women; moderate-quality evidence with significant heterogeneity). Quality-of-life scores were slightly better with bisphosphonates than placebo at comparable time points (in three out of five studies; moderate-quality evidence) however scores decreased during the course of the studies. Denosumab reduced the risk of developing a SRE compared with bisphosphonates by 22% (RR 0.78, 0.72 to 0.85; P < 0.001; 3 studies, 2345 women). One study reported data on overall survival and observed no difference in survival between denosumab and bisphosphonate.Reported toxicities across all settings were generally mild. Osteonecrosis of the jaw was rare, occurring less than 0.5% in the adjuvant setting (high-quality evidence). AUTHORS' CONCLUSIONS For women with EBC, bisphosphonates reduce the risk of bone metastases and provide an overall survival benefit compared to placebo or no bisphosphonates. There is preliminary evidence suggestive that bisphosphonates provide an overall survival and disease-free survival benefit in postmenopausal women only when compared to placebo or no bisphosphonate. This was not a planned subgroup for these early trials, and we await the completion of new large clinical trials assessing benefit for postmenopausal women. For women with BCBM, bisphosphonates reduce the risk of developing SREs, delay the median time to an SRE, and appear to reduce bone pain compared to placebo or no bisphosphonate.
Collapse
Affiliation(s)
- Brent O'Carrigan
- Chris O'Brien LifehouseMedical Oncology119‐143 Missenden RdCamperdownSydneyNSWUK2050
- The University of SydneyCamperdownAustralia
| | - Matthew HF Wong
- Gosford HospitalDepartment of Medical OncologyGosfordNew South WalesAustralia
| | - Melina L Willson
- NHMRC Clinical Trials Centre, The University of SydneySystematic Reviews and Health Technology AssessmentsLocked Bag 77SydneyNSWAustralia1450
| | - Martin R Stockler
- The University of SydneyNHMRC Clinical Trials Centre and Sydney Cancer CentreGH6 RPAHMissenden RoadCamperdownNSWAustralia2050
| | - Nick Pavlakis
- Royal North Shore HospitalDepartment of Medical OncologyPacific HighwaySt LeonardsNew South WalesAustralia2065
| | - Annabel Goodwin
- The University of Sydney, Concord Repatriation General HospitalConcord Clinical SchoolConcordNSWAustralia2137
- Concord Repatriation General HospitalMedical Oncology DepartmentConcordAustralia
- Sydney Local Health District and South Western Sydney Local Health DistrictCancer Genetics DepartmentSydneyAustralia
| | | |
Collapse
|
6
|
Dhesy-Thind S, Fletcher GG, Blanchette PS, Clemons MJ, Dillmon MS, Frank ES, Gandhi S, Gupta R, Mates M, Moy B, Vandenberg T, Van Poznak CH. Use of Adjuvant Bisphosphonates and Other Bone-Modifying Agents in Breast Cancer: A Cancer Care Ontario and American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 2017; 35:2062-2081. [PMID: 28618241 DOI: 10.1200/jco.2016.70.7257] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Purpose To make recommendations regarding the use of bisphosphonates and other bone-modifying agents as adjuvant therapy for patients with breast cancer. Methods Cancer Care Ontario and ASCO convened a Working Group and Expert Panel to develop evidence-based recommendations informed by a systematic review of the literature. Results Adjuvant bisphosphonates were found to reduce bone recurrence and improve survival in postmenopausal patients with nonmetastatic breast cancer. In this guideline, postmenopausal includes patients with natural menopause or that induced by ovarian suppression or ablation. Absolute benefit is greater in patients who are at higher risk of recurrence, and almost all trials were conducted in patients who also received systemic therapy. Most studies evaluated zoledronic acid or clodronate, and data are extremely limited for other bisphosphonates. While denosumab was found to reduce fractures, long-term survival data are still required. Recommendations It is recommended that, if available, zoledronic acid (4 mg intravenously every 6 months) or clodronate (1,600 mg/d orally) be considered as adjuvant therapy for postmenopausal patients with breast cancer who are deemed candidates for adjuvant systemic therapy. Further research comparing different bone-modifying agents, doses, dosing intervals, and durations is required. Risk factors for osteonecrosis of the jaw and renal impairment should be assessed, and any pending dental or oral health problems should be dealt with prior to starting treatment. Data for adjuvant denosumab look promising but are currently insufficient to make any recommendation. Use of these agents to reduce fragility fractures in patients with low bone mineral density is beyond the scope of the guideline. Recommendations are not meant to restrict such use of bone-modifying agents in these situations. Additional information at www.asco.org/breast-cancer-adjuvant-bisphosphonates-guideline , www.asco.org/guidelineswiki , https://www.cancercareontario.ca/guidelines-advice/types-of-cancer/breast .
Collapse
Affiliation(s)
- Sukhbinder Dhesy-Thind
- Sukhbinder Dhesy-Thind, Juravinski Cancer Centre; Sukhbinder Dhesy-Thind and Glenn G. Fletcher, McMaster University, Hamilton, Ontario; Phillip S. Blanchette, Sunnybrook Odette Cancer Centre; Sonal Gandhi, Sunnybrook Health Sciences, Toronto, Ontario; Mark J. Clemons, The Ottawa Hospital Cancer Centre, Ottawa, Ontario; Rasna Gupta, Windsor Regional Cancer Program, Windsor, Ontario; Mihaela Mates, Kingston General Hospital, Kingston, Ontario; Ted Vandenberg, London Health Sciences Centre, London, Ontario, Canada; Melissa S. Dillmon, Harbin Clinic, Rome, GA; Elizabeth S. Frank, Lexington; Beverly Moy, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; and Catherine H. Van Poznak, University of Michigan, Ann Arbor, MI
| | - Glenn G Fletcher
- Sukhbinder Dhesy-Thind, Juravinski Cancer Centre; Sukhbinder Dhesy-Thind and Glenn G. Fletcher, McMaster University, Hamilton, Ontario; Phillip S. Blanchette, Sunnybrook Odette Cancer Centre; Sonal Gandhi, Sunnybrook Health Sciences, Toronto, Ontario; Mark J. Clemons, The Ottawa Hospital Cancer Centre, Ottawa, Ontario; Rasna Gupta, Windsor Regional Cancer Program, Windsor, Ontario; Mihaela Mates, Kingston General Hospital, Kingston, Ontario; Ted Vandenberg, London Health Sciences Centre, London, Ontario, Canada; Melissa S. Dillmon, Harbin Clinic, Rome, GA; Elizabeth S. Frank, Lexington; Beverly Moy, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; and Catherine H. Van Poznak, University of Michigan, Ann Arbor, MI
| | - Phillip S Blanchette
- Sukhbinder Dhesy-Thind, Juravinski Cancer Centre; Sukhbinder Dhesy-Thind and Glenn G. Fletcher, McMaster University, Hamilton, Ontario; Phillip S. Blanchette, Sunnybrook Odette Cancer Centre; Sonal Gandhi, Sunnybrook Health Sciences, Toronto, Ontario; Mark J. Clemons, The Ottawa Hospital Cancer Centre, Ottawa, Ontario; Rasna Gupta, Windsor Regional Cancer Program, Windsor, Ontario; Mihaela Mates, Kingston General Hospital, Kingston, Ontario; Ted Vandenberg, London Health Sciences Centre, London, Ontario, Canada; Melissa S. Dillmon, Harbin Clinic, Rome, GA; Elizabeth S. Frank, Lexington; Beverly Moy, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; and Catherine H. Van Poznak, University of Michigan, Ann Arbor, MI
| | - Mark J Clemons
- Sukhbinder Dhesy-Thind, Juravinski Cancer Centre; Sukhbinder Dhesy-Thind and Glenn G. Fletcher, McMaster University, Hamilton, Ontario; Phillip S. Blanchette, Sunnybrook Odette Cancer Centre; Sonal Gandhi, Sunnybrook Health Sciences, Toronto, Ontario; Mark J. Clemons, The Ottawa Hospital Cancer Centre, Ottawa, Ontario; Rasna Gupta, Windsor Regional Cancer Program, Windsor, Ontario; Mihaela Mates, Kingston General Hospital, Kingston, Ontario; Ted Vandenberg, London Health Sciences Centre, London, Ontario, Canada; Melissa S. Dillmon, Harbin Clinic, Rome, GA; Elizabeth S. Frank, Lexington; Beverly Moy, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; and Catherine H. Van Poznak, University of Michigan, Ann Arbor, MI
| | - Melissa S Dillmon
- Sukhbinder Dhesy-Thind, Juravinski Cancer Centre; Sukhbinder Dhesy-Thind and Glenn G. Fletcher, McMaster University, Hamilton, Ontario; Phillip S. Blanchette, Sunnybrook Odette Cancer Centre; Sonal Gandhi, Sunnybrook Health Sciences, Toronto, Ontario; Mark J. Clemons, The Ottawa Hospital Cancer Centre, Ottawa, Ontario; Rasna Gupta, Windsor Regional Cancer Program, Windsor, Ontario; Mihaela Mates, Kingston General Hospital, Kingston, Ontario; Ted Vandenberg, London Health Sciences Centre, London, Ontario, Canada; Melissa S. Dillmon, Harbin Clinic, Rome, GA; Elizabeth S. Frank, Lexington; Beverly Moy, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; and Catherine H. Van Poznak, University of Michigan, Ann Arbor, MI
| | - Elizabeth S Frank
- Sukhbinder Dhesy-Thind, Juravinski Cancer Centre; Sukhbinder Dhesy-Thind and Glenn G. Fletcher, McMaster University, Hamilton, Ontario; Phillip S. Blanchette, Sunnybrook Odette Cancer Centre; Sonal Gandhi, Sunnybrook Health Sciences, Toronto, Ontario; Mark J. Clemons, The Ottawa Hospital Cancer Centre, Ottawa, Ontario; Rasna Gupta, Windsor Regional Cancer Program, Windsor, Ontario; Mihaela Mates, Kingston General Hospital, Kingston, Ontario; Ted Vandenberg, London Health Sciences Centre, London, Ontario, Canada; Melissa S. Dillmon, Harbin Clinic, Rome, GA; Elizabeth S. Frank, Lexington; Beverly Moy, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; and Catherine H. Van Poznak, University of Michigan, Ann Arbor, MI
| | - Sonal Gandhi
- Sukhbinder Dhesy-Thind, Juravinski Cancer Centre; Sukhbinder Dhesy-Thind and Glenn G. Fletcher, McMaster University, Hamilton, Ontario; Phillip S. Blanchette, Sunnybrook Odette Cancer Centre; Sonal Gandhi, Sunnybrook Health Sciences, Toronto, Ontario; Mark J. Clemons, The Ottawa Hospital Cancer Centre, Ottawa, Ontario; Rasna Gupta, Windsor Regional Cancer Program, Windsor, Ontario; Mihaela Mates, Kingston General Hospital, Kingston, Ontario; Ted Vandenberg, London Health Sciences Centre, London, Ontario, Canada; Melissa S. Dillmon, Harbin Clinic, Rome, GA; Elizabeth S. Frank, Lexington; Beverly Moy, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; and Catherine H. Van Poznak, University of Michigan, Ann Arbor, MI
| | - Rasna Gupta
- Sukhbinder Dhesy-Thind, Juravinski Cancer Centre; Sukhbinder Dhesy-Thind and Glenn G. Fletcher, McMaster University, Hamilton, Ontario; Phillip S. Blanchette, Sunnybrook Odette Cancer Centre; Sonal Gandhi, Sunnybrook Health Sciences, Toronto, Ontario; Mark J. Clemons, The Ottawa Hospital Cancer Centre, Ottawa, Ontario; Rasna Gupta, Windsor Regional Cancer Program, Windsor, Ontario; Mihaela Mates, Kingston General Hospital, Kingston, Ontario; Ted Vandenberg, London Health Sciences Centre, London, Ontario, Canada; Melissa S. Dillmon, Harbin Clinic, Rome, GA; Elizabeth S. Frank, Lexington; Beverly Moy, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; and Catherine H. Van Poznak, University of Michigan, Ann Arbor, MI
| | - Mihaela Mates
- Sukhbinder Dhesy-Thind, Juravinski Cancer Centre; Sukhbinder Dhesy-Thind and Glenn G. Fletcher, McMaster University, Hamilton, Ontario; Phillip S. Blanchette, Sunnybrook Odette Cancer Centre; Sonal Gandhi, Sunnybrook Health Sciences, Toronto, Ontario; Mark J. Clemons, The Ottawa Hospital Cancer Centre, Ottawa, Ontario; Rasna Gupta, Windsor Regional Cancer Program, Windsor, Ontario; Mihaela Mates, Kingston General Hospital, Kingston, Ontario; Ted Vandenberg, London Health Sciences Centre, London, Ontario, Canada; Melissa S. Dillmon, Harbin Clinic, Rome, GA; Elizabeth S. Frank, Lexington; Beverly Moy, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; and Catherine H. Van Poznak, University of Michigan, Ann Arbor, MI
| | - Beverly Moy
- Sukhbinder Dhesy-Thind, Juravinski Cancer Centre; Sukhbinder Dhesy-Thind and Glenn G. Fletcher, McMaster University, Hamilton, Ontario; Phillip S. Blanchette, Sunnybrook Odette Cancer Centre; Sonal Gandhi, Sunnybrook Health Sciences, Toronto, Ontario; Mark J. Clemons, The Ottawa Hospital Cancer Centre, Ottawa, Ontario; Rasna Gupta, Windsor Regional Cancer Program, Windsor, Ontario; Mihaela Mates, Kingston General Hospital, Kingston, Ontario; Ted Vandenberg, London Health Sciences Centre, London, Ontario, Canada; Melissa S. Dillmon, Harbin Clinic, Rome, GA; Elizabeth S. Frank, Lexington; Beverly Moy, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; and Catherine H. Van Poznak, University of Michigan, Ann Arbor, MI
| | - Ted Vandenberg
- Sukhbinder Dhesy-Thind, Juravinski Cancer Centre; Sukhbinder Dhesy-Thind and Glenn G. Fletcher, McMaster University, Hamilton, Ontario; Phillip S. Blanchette, Sunnybrook Odette Cancer Centre; Sonal Gandhi, Sunnybrook Health Sciences, Toronto, Ontario; Mark J. Clemons, The Ottawa Hospital Cancer Centre, Ottawa, Ontario; Rasna Gupta, Windsor Regional Cancer Program, Windsor, Ontario; Mihaela Mates, Kingston General Hospital, Kingston, Ontario; Ted Vandenberg, London Health Sciences Centre, London, Ontario, Canada; Melissa S. Dillmon, Harbin Clinic, Rome, GA; Elizabeth S. Frank, Lexington; Beverly Moy, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; and Catherine H. Van Poznak, University of Michigan, Ann Arbor, MI
| | - Catherine H Van Poznak
- Sukhbinder Dhesy-Thind, Juravinski Cancer Centre; Sukhbinder Dhesy-Thind and Glenn G. Fletcher, McMaster University, Hamilton, Ontario; Phillip S. Blanchette, Sunnybrook Odette Cancer Centre; Sonal Gandhi, Sunnybrook Health Sciences, Toronto, Ontario; Mark J. Clemons, The Ottawa Hospital Cancer Centre, Ottawa, Ontario; Rasna Gupta, Windsor Regional Cancer Program, Windsor, Ontario; Mihaela Mates, Kingston General Hospital, Kingston, Ontario; Ted Vandenberg, London Health Sciences Centre, London, Ontario, Canada; Melissa S. Dillmon, Harbin Clinic, Rome, GA; Elizabeth S. Frank, Lexington; Beverly Moy, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA; and Catherine H. Van Poznak, University of Michigan, Ann Arbor, MI
| |
Collapse
|
7
|
O'Bryant SE, Mielke MM, Rissman RA, Lista S, Vanderstichele H, Zetterberg H, Lewczuk P, Posner H, Hall J, Johnson L, Fong YL, Luthman J, Jeromin A, Batrla-Utermann R, Villarreal A, Britton G, Snyder PJ, Henriksen K, Grammas P, Gupta V, Martins R, Hampel H. Blood-based biomarkers in Alzheimer disease: Current state of the science and a novel collaborative paradigm for advancing from discovery to clinic. Alzheimers Dement 2017; 13:45-58. [PMID: 27870940 PMCID: PMC5218961 DOI: 10.1016/j.jalz.2016.09.014] [Citation(s) in RCA: 206] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 09/27/2016] [Indexed: 11/25/2022]
Abstract
The last decade has seen a substantial increase in research focused on the identification of blood-based biomarkers that have utility in Alzheimer's disease (AD). Blood-based biomarkers have significant advantages of being time- and cost-efficient as well as reduced invasiveness and increased patient acceptance. Despite these advantages and increased research efforts, the field has been hampered by lack of reproducibility and an unclear path for moving basic discovery toward clinical utilization. Here we reviewed the recent literature on blood-based biomarkers in AD to provide a current state of the art. In addition, a collaborative model is proposed that leverages academic and industry strengths to facilitate the field in moving past discovery only work and toward clinical use. Key resources are provided. This new public-private partnership model is intended to circumvent the traditional handoff model and provide a clear and useful paradigm for the advancement of biomarker science in AD and other neurodegenerative diseases.
Collapse
Affiliation(s)
- Sid E O'Bryant
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA.
| | - Michelle M Mielke
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Department of Health Science Research, Mayo Clinic, Rochester, MN, USA
| | - Robert A Rissman
- Alzheimer's Disease Cooperative Study, Department of Neurosciences, UCSD School of Medicine, La Jolla, CA, USA
| | - Simone Lista
- AXA Research Fund and UPMC Chair, Paris, France; Department de Neurologie, Institut de la Memorie et de la Maladie d'Alzheimer (IM2A) et Institut du Cerveau et du la Moelle epiniere (ICM), Hospital de la Pitie-Salpetriere, Sorbonne Universites, Universite Pierre et Marie Curie, Paris, France
| | | | - Henrik Zetterberg
- Clinical Neurochemistry Laboratory, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gotenburg, Molndal, Sweden; UCL Institute of Neurology, London, UK
| | - Piotr Lewczuk
- Department of Psychiatry and Psychotherapy, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany; Department of Neurodegeneration Diagnostics, Medical University of Bialystok, Bialystok, Poland
| | | | - James Hall
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Leigh Johnson
- Institute for Healthy Aging, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Yiu-Lian Fong
- Johnson & Johnson, London Innovation Center, London, UK
| | - Johan Luthman
- Neuroscience Clinical Development, Clinical Neuroscience Eisai, Woodcliff Lake, NJ, USA
| | | | | | - Alcibiades Villarreal
- Centro de Neurociencias y Unidad de Investigacion Clinica, Instituto de Investigaciones Cientificas y Servicios de Alta Tecnologia (INDICASAT AIP), Ciudad del Saber, Panama, Panama
| | - Gabrielle Britton
- Centro de Neurociencias y Unidad de Investigacion Clinica, Instituto de Investigaciones Cientificas y Servicios de Alta Tecnologia (INDICASAT AIP), Ciudad del Saber, Panama, Panama
| | - Peter J Snyder
- Department of Neurology, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA
| | - Kim Henriksen
- Neurodegenerative Diseases, Nordic Bioscience Biomarkers and Research, Herlev, Denmark
| | - Paula Grammas
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, RI, USA
| | - Veer Gupta
- Faculty of Health, Engineering and Sciences, Center of Excellence for Alzheimer's Disease Research and Care, School of Medical Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Ralph Martins
- Faculty of Health, Engineering and Sciences, Center of Excellence for Alzheimer's Disease Research and Care, School of Medical Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Harald Hampel
- AXA Research Fund and UPMC Chair, Paris, France; Department de Neurologie, Institut de la Memorie et de la Maladie d'Alzheimer (IM2A) et Institut du Cerveau et du la Moelle epiniere (ICM), Hospital de la Pitie-Salpetriere, Sorbonne Universites, Universite Pierre et Marie Curie, Paris, France
| |
Collapse
|
8
|
Reproductive hormones in breast cancer bone metastasis: The role of inhibins. J Bone Oncol 2016; 5:139-142. [PMID: 27761376 PMCID: PMC5063224 DOI: 10.1016/j.jbo.2016.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 03/04/2016] [Accepted: 03/07/2016] [Indexed: 11/21/2022] Open
Abstract
The spread of breast cancer cells to bone and survival in this new metastatic environment is influenced not only by the genetic signature of the cells, but also multiple host cells and soluble factors produced locally (paracrine) or from distant sites (endocrine). Disrupting this metastatic process has been evaluated in clinical trials of the bone targeted agents bisphosphonates and denosumab and have shown that these agents reduce the recurrence of breast cancer in postmenopausal women only, suggesting the efficacy of the drugs are influenced by levels of reproductive endocrine hormones. The molecular mechanism driving this differential effect has not been definitively identified, however, there is evidence that both reproductive hormones and bisphosphonates can affect similar paracrine factors and cellular components of the bone metastatic niche. This review focuses on how the ovarian endocrine hormone, inhibin, interacts with the paracrine factors activin and follistatin, abundant in the primary tumour and bone microenvironment, with subsequent effects on tumour cell survival. Inhibin also affects the cellular components of the bone microenvironment primarily the osteoblastic niche. Recent evidence has shown that bisphosphonates also alter this niche, which may represent a common mechanism by which inhibin and bisphosphonates interact to influence disease outcomes in early breast cancer. Further research is needed to fully elucidate these molecular mechanisms to enable understanding and future development of alternative bone targeted treatments with anti-tumour efficacy in premenopausal women.
Collapse
|
9
|
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: 2.8] [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
|
10
|
Van Acker HH, Anguille S, Willemen Y, Smits EL, Van Tendeloo VF. Bisphosphonates for cancer treatment: Mechanisms of action and lessons from clinical trials. Pharmacol Ther 2015; 158:24-40. [PMID: 26617219 DOI: 10.1016/j.pharmthera.2015.11.008] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A growing body of evidence points toward an important anti-cancer effect of bisphosphonates, a group of inexpensive, safe, potent, and long-term stable pharmacologicals that are widely used as osteoporosis drugs. To date, they are already used in the prevention of complications of bone metastases. Because the bisphosphonates can also reduce mortality in among other multiple myeloma, breast, and prostate cancer patients, they are now thoroughly studied in oncology. In particular, the more potent nitrogen-containing bisphosphonates have the potential to improve prognosis. The first part of this review will elaborate on the direct and indirect anti-tumoral effects of bisphosphonates, including induction of tumor cell apoptosis, inhibition of tumor cell adhesion and invasion, anti-angiogenesis, synergism with anti-neoplastic drugs, and enhancement of immune surveillance (e.g., through activation of γδ T cells and targeting macrophages). In the second part, we shed light on the current clinical position of bisphosphonates in the treatment of hematological and solid malignancies, as well as on ongoing and completed clinical trials investigating the therapeutic effect of bisphosphonates in cancer. Based on these recent data, the role of bisphosphonates is expected to further expand in the near future outside the field of osteoporosis and to open up new avenues in the treatment of malignancies.
Collapse
Affiliation(s)
- Heleen H Van Acker
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium.
| | - Sébastien Anguille
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Yannick Willemen
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Evelien L Smits
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium; Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium; Center for Oncological Research (CORE), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Viggo F Van Tendeloo
- Laboratory of Experimental Hematology, Tumor Immunology Group (TIGR), Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
11
|
Neoadjuvant treatment of breast cancer: maximizing pathologic complete response rates to improve prognosis. Curr Opin Obstet Gynecol 2015; 27:85-91. [PMID: 25490376 DOI: 10.1097/gco.0000000000000147] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW Neoadjuvant therapy is very often an adequate alternative to adjuvant therapy. This review summarizes the recent advances made in the area of neoadjuvant therapy in breast cancer. The focus will lie on recently published clinical trials, but will not further highlight surgical, imaging and radio-oncological issues related to neoadjuvant therapy. RECENT FINDINGS Within the past year, it has been discussed if neoadjuvant treatment can be used as a faster way to get access to new therapies, based on new data in HER2+ breast cancer, suggesting a higher pathological complete response rate when a dual anti-HER2 therapy was used. Nevertheless, this higher pathological complete response rate does not necessarily always translate into a better survival. In triple negative breast cancer, carboplatin could be identified as an asset for patients, especially in patients with gBRCA mutations. However, mature long-term data are still missing. The neoadjuvant approach is ideal to identify new biomarkers which predict response or resistance to the given treatment. Tumour-infiltrating lymphocytes and PIK3CA mutations are amongst the most promising markers. SUMMARY Neoadjuvant treatment should be considered for all patients with HER2-positive or triple negative breast cancer. Clinical trials in this setting are currently investigating new approaches.
Collapse
|
12
|
Addison CL, Pond GR, Cochrane B, Zhao H, Chia SK, Levine MN, Clemons M. Correlation of baseline biomarkers with clinical outcomes and response to fulvestrant with vandetanib or placebo in patients with bone predominant metastatic breast cancer: An OCOG ZAMBONEY sub-study. J Bone Oncol 2015; 4:47-53. [PMID: 26579488 PMCID: PMC4620970 DOI: 10.1016/j.jbo.2015.04.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Revised: 04/16/2015] [Accepted: 04/17/2015] [Indexed: 12/14/2022] Open
Abstract
Background Bone metastases are common in women with breast cancer and often result in skeletal related events (SREs). As the angiogenic factor vascular endothelial growth factor (VEGF) regulates osteoclast activity and is associated with more extensive bone metastases and SRE risk in metastatic breast cancer, we hypothesized that blockade of VEGF signaling could be a therapeutic strategy for inhibiting bone metastases progression and possibly prolonging overall (OS) or progression-free survival (PFS). The Zamboney trial was a randomized placebo-controlled study designed to assess whether patients with bone predominant metastatic breast cancer benefited from addition of the VEGF receptor (VEGFR) targeting agent, vandetanib, to endocrine therapy with fulvestrant. As a companion study, evaluation of biomarkers and their potential association with response to vandetanib or SRE risk was performed. Methods Baseline overnight fasted serum from enrolled patients was analyzed for levels of various putative biomarkers including; VEGF-A, soluble (s)VEGFR2, sVEGFR3, transforming growth factor (TGF)-β1 and activinA by ELISA. Spearman correlation coefficients and Wilcoxon rank sum tests were used to investigate potential relationships between biomarker values and baseline clinical parameters. Prognostic and predictive ability of each marker was investigated using Cox proportional hazards regression with adjustments for treatment and baseline strata of serum CTx (<400 versus ≥400 ng/L). Results Of 129 enrolled patients, serum was available for analysis in 101; 51 in vandetanib and 50 in placebo arm. Mean age amongst consenting patients was 59.8 years. Clinical characteristics were not significantly different between patients with or without serum biomarker data and serum markers were similar for patients by treatment arm. Baseline sVEGFR2 was prognostic for OS (HR=0.77, 95% CI=0.61–0.96, p=0.020), and although a modest association was observed, it was not significant for PFS (HR=0.90, 95% CI=0.80–1.01, p=0.085) nor time to first SRE (HR=0.82, 95% CI=0.66–1.02, p=0.079). When interaction terms were evaluated, sVEGFR2 was not found to be predictive of response to vandetanib, although a modest association remained with respect to PFS (interaction p=0.085). No other marker showed any significant prognostic or predictive ability with any measured outcome. Conclusions In this clinical trial, sVEGFR2 appeared prognostic for OS, hence validation of sVEGFR2 should be conducted. Moreover, the role of sVEGFR2 in breast cancer bone metastasis progression should be elucidated. Baseline VEGF, sVEGFR3, TGF-β or activinA were not associated with clinical outcomes in patients treated with fulvestrant in conjunction with vandetanib or placebo. Baseline sVEGFR2 was modestly associated with clinical outcomes including PFS, OS and time to first skeletal event. Increased baseline sVEGFR2 was associated with improved clinical outcomes in this study sample. These findings support the need for future studies of the role of sVEGFR2 in bone metastasis progression.
Collapse
Key Words
- BP, bisphosphonate
- BPI, brief pain inventory
- Biomarker
- Bone metastasis
- Breast cancer
- CTx, C-telopeptide
- ER, estrogen receptor
- FACT-BP, Functional assessment of cancer therapy-bone pain
- OS, overall survival
- PFS, progression free survival
- PR, progesterone receptor
- Patient outcome
- RANKL, Receptor Activator NF-KB ligand
- SRE, skeletal related event
- Skeletal related event
- TGF-β, transforming growth factor beta
- VEGF, vascular endothelial growth factor
- Vandetanib
- sVEGFR, soluble vascular endothelial growth factor receptor
- uNTx, urinary N-telopeptide
Collapse
Affiliation(s)
- Christina L Addison
- Program for Cancer Therapeutics, Ottawa Hospital Research Institute, Box 926, 501 Smyth Road, Ottawa, ON, Canada K1H 8L6 ; Department of Medicine, University of Ottawa, Ottawa, ON, Canada ; Department of Biochemistry, Microbiology and Immunology University of Ottawa, Ottawa, ON, Canada
| | - Gregory R Pond
- McMaster University and Ontario Clinical Oncology Group, Hamilton, ON, Canada
| | - Brandy Cochrane
- McMaster University and Ontario Clinical Oncology Group, Hamilton, ON, Canada
| | - Huijun Zhao
- Program for Cancer Therapeutics, Ottawa Hospital Research Institute, Box 926, 501 Smyth Road, Ottawa, ON, Canada K1H 8L6
| | | | - Mark N Levine
- McMaster University and Ontario Clinical Oncology Group, Hamilton, ON, Canada
| | - Mark Clemons
- Program for Cancer Therapeutics, Ottawa Hospital Research Institute, Box 926, 501 Smyth Road, Ottawa, ON, Canada K1H 8L6 ; Department of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
13
|
A randomised controlled trial of intravenous zoledronic acid in malignant pleural disease: a proof of principle pilot study. PLoS One 2015; 10:e0118569. [PMID: 25781025 PMCID: PMC4364455 DOI: 10.1371/journal.pone.0118569] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Accepted: 01/20/2015] [Indexed: 12/29/2022] Open
Abstract
Introduction Animal studies have shown Zoledronic Acid (ZA) may diminish pleural fluid accumulation and tumour bulk in malignant pleural disease (MPD). We performed a pilot study to evaluate its effects in humans. Methods We undertook a single centre, double-blind, placebo-controlled trial in adults with MPD. Patients were randomised (1:1) to receive 2 doses of intravenous ZA or placebo, 3 weeks apart and were followed-up for 6 weeks. The co-primary outcomes were change in Visual Analogue Scale (VAS) score measured breathlessness during trial follow-up and change in the initial area under the curve (iAUC) on thoracic Dynamic Contrast Enhanced Magnetic Resonance Imaging (DCE-MRI) from randomisation to week 5. Multiple secondary endpoints were also evaluated. Results Between January 2010 and May 2013, 30 patients were enrolled, 24 randomised and 4 withdrew after randomisation (1 withdrew consent; 3 had a clinical decline). At baseline, the ZA group were more breathless, had more advanced disease on radiology and worse quality of life than the placebo group. There was no significant difference between the groups with regards change in breathlessness (Adjusted mean difference (AMD) 4.16 (95%CI −4.7 to 13.0)) or change in DCE-MRI iAUC (AMD −15.4 (95%CI −58.1 to 27.3). Two of nine (22%) in the ZA arm had a >10% improvement by modified RECIST (vs 0/11 who received placebo). There was no significant difference in quality of life measured by the QLQ-C30 score (global QOL: AMD -4.1 (-13.0 to 4.9)), side effects or serious adverse event rates. Conclusions This is the first human study to evaluate ZA in MPD. The study is limited by small numbers and imbalanced baseline characteristics. Although no convincing treatment effect was identified, potential benefits for specific subgroups of patients cannot be excluded. This study provides important information regarding the feasibility of future trials to evaluate the effects of ZA further. Trial Registration UK Clinical Research Network ID 8877 ISRCTN17030426www.isrctn.com
Collapse
|
14
|
Wilson C, Ottewell P, Coleman RE, Holen I. The differential anti-tumour effects of zoledronic acid in breast cancer - evidence for a role of the activin signaling pathway. BMC Cancer 2015; 15:55. [PMID: 25884855 PMCID: PMC4329195 DOI: 10.1186/s12885-015-1066-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 02/02/2015] [Indexed: 12/20/2022] Open
Abstract
Background Neo-adjuvant breast cancer clinical trials of zoledronic acid (ZOL) have shown that patients with oestrogen negative (ER-ve) tumours have improved disease outcomes. We investigated the molecular mechanism behind this differential anti-tumour effect according to ER status, hypothesising it may in part be mediated via the activin signaling pathway. Methods The effects of activin A, its inhibitor follistatin and zoledronic acid on proliferation of breast cancer cells was evaluated using either an MTS proliferation assay or trypan blue. Secretion of activin A and follistatin in conditioned medium (CM) from MDA-MB-231, MDA-MB-436, MCF7 and T47D cell lines were measured using specific ELISAs. The effects of ZOL on phosphorylation domains of Smad2 (pSmad2c + pSmad2L) were evaluated using immunofluorescence. Changes seen in vitro were confirmed in a ZOL treated subcutaneous ER-ve MDA-MB-436 xenograft model. Results Activin A inhibits proliferation of both ER-ve and oestrogen positive (ER + ve) breast cancer cells, an effect impaired by follistatin. ZOL significantly inhibits proliferation and the secretion of follistatin from ER-ve cells only, which increases the biological activity of the canonical activin A pathway by significantly increasing intracellular pSmad2c and decreasing nuclear accumulation of pSmad2L. In vivo, ZOL significantly decreases follistatin and pSmad2L expression in ER-ve subcutaneous xenografts compared to saline treated control animals. Conclusions This is the first report showing a differential effect of ZOL, according to ER status, on the activin pathway and its inhibitors in vitro and in vivo. These data suggest a potential molecular mechanism contributing to the differential anti-tumour effects reported from clinical trials and requires further evaluation in clinical samples.
Collapse
Affiliation(s)
- Caroline Wilson
- Academic Unit of Clinical Oncology, University of Sheffield, Medical School, Sheffield, UK.
| | - Penelope Ottewell
- Academic Department of Oncology, University of Sheffield, Medical School, Sheffield, UK.
| | - Robert E Coleman
- Academic Unit of Clinical Oncology, University of Sheffield, Medical School, Sheffield, UK.
| | - Ingunn Holen
- Academic Unit of Clinical Oncology, University of Sheffield, Medical School, Sheffield, UK.
| |
Collapse
|
15
|
Prinsloo S, Wei Q, Scott SM, Tannir N, Jonasc E, Pisters L, Cohen L. Psychological states, serum markers and survival: associations and predictors of survival in patients with renal cell carcinoma. J Behav Med 2015; 38:48-56. [PMID: 24935017 PMCID: PMC4824635 DOI: 10.1007/s10865-014-9578-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 05/31/2014] [Indexed: 11/26/2022]
Abstract
This study sought to determine if there was an association between prognostic-based serum biomarkers, survival, and psychosocial factors in patients with metastatic renal cell carcinoma. Associations were found between psychosocial factors and biomarker levels (hemoglobin with depressive symptoms (r = -0.29), positive affect (r = 0.30), social support (r = 0.19), and perceived stress (r = -0.27); albumin with depressive symptoms (r = -0.19), positive affect (r = 0.22), and social support (r = 0.20); alkaline phosphatase with depressive symptoms (r = 0.21), all p values <0.05. After adjustment for disease-related risk factors, only the associations between positive affect and perceived stress with hemoglobin remained significant (p's < 0.05). Positive affect (HR = 0.90; 95% CI = 0.83, 0.97; p = 0.009) and depressive symptom total scores (HR = 1.03; 95% CI = 1.01, 1.06; p = 0.013), and alkaline phosphatase (HR 2.72; 95% CI = 1.41, 5.24; p = 0.003) were associated with survival. This study suggests that measures of positive and negative psychological outlook may contribute differently to health, well-being, and survival.
Collapse
Affiliation(s)
- Sarah Prinsloo
- Unit 410, Department of General Oncology and the Integrative Medicine Program, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77230, USA
| | - Qi Wei
- Unit 410, Department of General Oncology and the Integrative Medicine Program, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77230, USA
| | - Shellie M. Scott
- Department of Urology, University of Texas MD Anderson, Cancer Center, Houston, TX, USA
| | - Nizar Tannir
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eric Jonasc
- Department of Genitourinary Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Louis Pisters
- Department of Urology, University of Texas MD Anderson, Cancer Center, Houston, TX, USA
| | - Lorenzo Cohen
- Unit 410, Department of General Oncology and the Integrative Medicine Program, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77230, USA
| |
Collapse
|
16
|
Roberts JN, Karvonen C, Graham K, Weinfeld M, Joy AA, Koebel M, Morris D, Robson PJ, Johnston RN, Brockton NT. Biobanking in the Twenty-First Century: Driving Population Metrics into Biobanking Quality. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 864:95-114. [DOI: 10.1007/978-3-319-20579-3_8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
17
|
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
|
18
|
Prevention of Bone Metastases in Breast Cancer Patients. Therapeutic Perspectives. J Clin Med 2014; 3:521-36. [PMID: 26237389 PMCID: PMC4449698 DOI: 10.3390/jcm3020521] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Revised: 03/03/2014] [Accepted: 03/14/2014] [Indexed: 01/13/2023] Open
Abstract
One in four breast cancer patients is at risk of developing bone metastases in her life time. The early prevention of bone metastases is a crucial challenge. It has been suggested that the use of zoledronic acid (ZOL) in the adjuvant setting may reduce the persistence of disseminated tumor cells and thereby might improve outcome, specifically in a population of patients with a low estrogen microenvironment. More recently, the results of a large meta-analysis from 41 randomized trials comparing a bisphosphonate (BP) to placebo or to an open control have been presented at the 2013 San Antonio Breast Cancer Meeting. Data on 17,016 patients confirm that adjuvant BPs, irrespective of the type of treatment or the treatment schedule and formulation (oral or intra-venously (IV)), significantly reduced bone recurrences and improved breast cancer survival in postmenopausal women. No advantage was seen in premenopausal women. BPs are soon likely to become integrated into standard practice. Published data on the mechanisms involved in tumor cell seeding from the primary site, in homing to bone tissues and in the reactivation of dormant tumor cells will be reviewed; these might offer new ideas for innovative combination strategies.
Collapse
|
19
|
Zekri J, Mansour M, Karim SM. The anti-tumour effects of zoledronic acid. J Bone Oncol 2014; 3:25-35. [PMID: 26909294 PMCID: PMC4723416 DOI: 10.1016/j.jbo.2013.12.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Revised: 12/16/2013] [Accepted: 12/19/2013] [Indexed: 11/20/2022] Open
Abstract
Bone is the most common site for metastasis in patients with solid tumours. Bisphosphonates are an effective treatment for preventing skeletal related events and preserving quality of life in these patients. Zoledronic acid (ZA) is the most potent osteoclast inhibitor and is licensed for the treatment of bone metastases. Clodronate and pamidronate are also licensed for this indication. In addition, ZA has been demonstrated to exhibit antitumour effect. Direct and indirect mechanisms of anti-tumour effect have been postulated and at many times proven. Evidence exists that ZA antitumour effect is mediated through inhibition of tumour cells proliferation, induction of apoptosis, synergistic/additive to inhibitory effect of cytotoxic agents, inhibition of angiogenesis, decrease tumour cells adhesion to bone, decrease tumour cells invasion and migration, disorganization of cell cytoskeleton and activation of specific cellular antitumour immune response. There is also clinical evidence from clinical trials that ZA improved long term survival outcome in cancer patients with and without bone metastases. In this review we highlight the preclinical and clinical studies investigating the antitumour effect of bisphosphonates with particular reference to ZA.
Collapse
Affiliation(s)
- Jamal Zekri
- AlFaisal University College of Medicine, PO Box 50927, Riyadh 11533, Saudi Arabia
- King Faisal Specilalist Hospital and Research Center, MBC J64, PO Box 40047, Jeddah 21499, Saudi Arabia
| | - Maged Mansour
- Jeddah Cancer Centre, Dr. Erfan & Bagedo General Hospital, King Fahd Street, PO Box 6519, Jeddah 21452, Saudi Arabia
| | - Syed Mustafa Karim
- AlFaisal University College of Medicine, PO Box 50927, Riyadh 11533, Saudi Arabia
- King Faisal Specilalist Hospital and Research Center, MBC J64, PO Box 40047, Jeddah 21499, Saudi Arabia
| |
Collapse
|
20
|
Foroni C, Milan M, Strina C, Cappelletti M, Fumarola C, Bonelli M, Bertoni R, Ferrero G, Maldotti M, Takano E, Andreis D, Venturini S, Brugnoli G, Petronini PG, Zanoni V, Pritzker L, Pritzker K, Parissenti A, Santini D, Fox SB, Bottini A, Generali D. Pure anti-tumor effect of zoledronic acid in naïve bone-only metastatic and locally advanced breast cancer: proof from the “biological window therapy”. Breast Cancer Res Treat 2014; 144:113-21. [DOI: 10.1007/s10549-014-2840-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Accepted: 01/11/2014] [Indexed: 12/14/2022]
|
21
|
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
|
22
|
Coleman R, Gregory W, Marshall H, Wilson C, Holen I. The metastatic microenvironment of breast cancer: Clinical implications. Breast 2013; 22 Suppl 2:S50-6. [DOI: 10.1016/j.breast.2013.07.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
|
23
|
Regulation of VEGF by mevalonate pathway inhibition in breast cancer. J Bone Oncol 2013; 2:110-5. [PMID: 26909280 PMCID: PMC4723384 DOI: 10.1016/j.jbo.2013.05.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 05/28/2013] [Accepted: 05/31/2013] [Indexed: 11/22/2022] Open
Abstract
Aminobisphosphonates are used for the treatment of benign and malignant bone disorders. As inhibitors of the mevalonate pathway they exert direct anti-tumor effects in vitro and in preclinical models of bone metastases. Bisphosphonates are thought to have an anti-angiogenic activity as decreased levels of VEGF have been reported in some, although not all patients, following treatment with bisphosphonates. Direct effects of bisphosphonates on tumor derived VEGF have not been examined in detail. We therefore investigated VEGF expression in breast cancer cell lines following mevalonate pathway inhibition. Treatment of cell lines with increasing doses of zoledronic acid and atorvastatin resulted in increased levels of VEGF production. Similar results were seen with the geranylgeranyltransferase I inhibitor GGTI-298. The induction of VEGF was reversed by the supplementation of geranylgeranyl pyrophosphate but not by farnesyl pyrophosphate indicating that this effect is mediated by inhibited geranylgeranylation. Previous reports have reported decreased VEGF levels in patients following BP treatment in vivo. We assessed VEGF levels in patients with non-metastatic breast cancer following repeated treatment with zoledronic acid. In contrast to our in vitro findings, VEGF serum levels decreased in all patients after 6–9 months of treatment (by an average of 41%) as assessed in a small pilot trial. These results indicate that tissues other than breast tumors contribute to the serum pool of circulating VEGF and may be responsible for the observed VEGF decreases. The increases of VEGF in the cancer cells may provide a rationale for the combined treatment with VEGF inhibitors.
Collapse
|