51
|
Kaliya-Perumal AK, Lu ML, Luo CA, Tsai TT, Lai PL, Chen LH, Chen WJ, Niu CC. Retrospective radiological outcome analysis following teriparatide use in elderly patients undergoing multilevel instrumented lumbar fusion surgery. Medicine (Baltimore) 2017; 96:e5996. [PMID: 28151894 PMCID: PMC5293457 DOI: 10.1097/md.0000000000005996] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Elderly patients with chronic nonresolving symptoms due to degenerative spine pathologies are prone to have poor surgical outcomes and hardware-related complications, especially following multilevel instrumented lumbar fusion surgeries. With intention of analyzing if teriparatide can be an effective adjunct therapy to surgical management, radiological outcomes are studied. Sixty-two elderly patients were divided into 2 similar groups. Group 1 (n = 30; mean age = 69.83 years; fusion levels = 137; screws = 269) had taken teriparatide (20 mcg SC injection, once daily) for a duration of 7.4 ± 2.4 months following surgery and Group 2 (n = 32; mean age = 70.38 years; fusion levels = 144; screws = 283) did not take teriparatide. Radiological evaluation was done to determine the state of postero lateral fusion mass and to investigate the incidence of pedicle screw loosening at 1-year follow-up. Unilateral or bilateral bridging bone formation across the transverse process of adjacent vertebras showing continuous trabeculation suggestive of solid fusion was obtained in 66.7% patients in the teriparatide group and 50% patients in the control group (P = 0.20). 13.4% of the total no. of screws showed signs of loosening in the teriparatide group, compared to 24.4% in the control group (P = 0.001). Percentage of patients achieving solid fusion following teriparatide use was found to be more than that of the control group. This difference may have clinical importance but was not statistically significant. However, teriparatide use was more significantly influential in reducing the incidence of subsequent pedicle screw loosening.
Collapse
Affiliation(s)
- Arun-Kumar Kaliya-Perumal
- Department of Orthopaedic Surgery, Spine Division, Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Department of Orthopaedic Surgery, Melmaruvathur Adhiparasakthi Institute of Medical Sciences and Research, Melmaruvathur, Tamil Nadu, India
| | - Meng-Ling Lu
- Department of Orthopaedic Surgery, Spine Division, Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chi-An Luo
- Department of Orthopaedic Surgery, Spine Division, Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Tsung-Ting Tsai
- Department of Orthopaedic Surgery, Spine Division, Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Po-Liang Lai
- Department of Orthopaedic Surgery, Spine Division, Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Lih-Huei Chen
- Department of Orthopaedic Surgery, Spine Division, Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wen-Jer Chen
- Department of Orthopaedic Surgery, Spine Division, Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chi-Chien Niu
- Department of Orthopaedic Surgery, Spine Division, Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
52
|
Martin TJ, Seeman E. Abaloparatide Is an Anabolic, but Does It Spare Resorption? J Bone Miner Res 2017; 32:11-16. [PMID: 27859635 DOI: 10.1002/jbmr.3042] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/08/2016] [Accepted: 11/15/2016] [Indexed: 01/04/2023]
Affiliation(s)
- T John Martin
- St Vincent's Institute of Medical Research, Melbourne, Australia
- Department of Medicine, St Vincent's Health, University of Melbourne, Melbourne, Australia
| | - Ego Seeman
- Department of Medicine, Austin Health, University of Melbourne, Melbourne, Australia
- Institute of Health and Ageing, Australian Catholic University, Melbourne, Australia
| |
Collapse
|
53
|
Osagie-Clouard L, Sanghani A, Coathup M, Briggs T, Bostrom M, Blunn G. Parathyroid hormone 1-34 and skeletal anabolic action: The use of parathyroid hormone in bone formation. Bone Joint Res 2017; 6:14-21. [PMID: 28062525 PMCID: PMC5227055 DOI: 10.1302/2046-3758.61.bjr-2016-0085.r1] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 10/24/2016] [Indexed: 12/19/2022] Open
Abstract
Intermittently administered parathyroid hormone (PTH 1-34) has been shown to promote bone formation in both human and animal studies. The hormone and its analogues stimulate both bone formation and resorption, and as such at low doses are now in clinical use for the treatment of severe osteoporosis. By varying the duration of exposure, parathyroid hormone can modulate genes leading to increased bone formation within a so-called 'anabolic window'. The osteogenic mechanisms involved are multiple, affecting the stimulation of osteoprogenitor cells, osteoblasts, osteocytes and the stem cell niche, and ultimately leading to increased osteoblast activation, reduced osteoblast apoptosis, upregulation of Wnt/β-catenin signalling, increased stem cell mobilisation, and mediation of the RANKL/OPG pathway. Ongoing investigation into their effect on bone formation through 'coupled' and 'uncoupled' mechanisms further underlines the impact of intermittent PTH on both cortical and cancellous bone. Given the principally catabolic actions of continuous PTH, this article reviews the skeletal actions of intermittent PTH 1-34 and the mechanisms underlying its effect. CITE THIS ARTICLE L. Osagie-Clouard, A. Sanghani, M. Coathup, T. Briggs, M. Bostrom, G. Blunn. Parathyroid hormone 1-34 and skeletal anabolic action: The use of parathyroid hormone in bone formation. Bone Joint Res 2017;6:14-21. DOI: 10.1302/2046-3758.61.BJR-2016-0085.R1.
Collapse
Affiliation(s)
- L Osagie-Clouard
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - A Sanghani
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - M Coathup
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - T Briggs
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - M Bostrom
- Hospital for Special Surgery, New York, New York, USA
| | - G Blunn
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| |
Collapse
|
54
|
Vrahnas C, Pearson TA, Brunt AR, Forwood MR, Bambery KR, Tobin MJ, Martin TJ, Sims NA. Anabolic action of parathyroid hormone (PTH) does not compromise bone matrix mineral composition or maturation. Bone 2016; 93:146-154. [PMID: 27686599 DOI: 10.1016/j.bone.2016.09.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 09/23/2016] [Accepted: 09/25/2016] [Indexed: 02/04/2023]
Abstract
Intermittent administration of parathyroid hormone (PTH) is used to stimulate bone formation in patients with osteoporosis. A reduction in the degree of matrix mineralisation has been reported during treatment, which may reflect either production of undermineralised matrix or a greater proportion of new matrix within the bone samples assessed. To explore these alternatives, high resolution synchrotron-based Fourier Transform Infrared Microspectroscopy (sFTIRM) coupled with calcein labelling was used in a region of non-remodelling cortical bone to determine bone composition during anabolic PTH treatment compared with region-matched samples from controls. 8week old male C57BL/6 mice were treated with vehicle or 50μg/kg PTH, 5 times/week for 4weeks (n=7-9/group). Histomorphometry confirmed greater trabecular and periosteal bone formation and 3-point bending tests confirmed greater femoral strength in PTH-treated mice. Dual calcein labels were used to match bone regions by time-since-mineralisation (bone age) and composition was measured by sFTIRM in six 15μm2 regions at increasing depth perpendicular to the most immature bone on the medial periosteal edge; this allowed in situ measurement of progressive changes in bone matrix during its maturation. The sFTIRM method was validated in vehicle-treated bones where the expected progressive increases in mineral:matrix ratio and collagen crosslink type ratio were detected with increasing bone maturity. We also observed a gradual increase in carbonate content that strongly correlated with an increase in longitudinal stretch of the collagen triple helix (amide I:amide II ratio). PTH treatment did not alter the progressive changes in any of these parameters from the periosteal edge through to the more mature bone. These data provide new information about how the bone matrix matures in situ and confirm that bone deposited during PTH treatment undergoes normal collagen maturation and normal mineral accrual.
Collapse
Affiliation(s)
- Christina Vrahnas
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; The University of Melbourne, Department of Medicine at St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Thomas A Pearson
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; The University of Melbourne, Department of Medicine at St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Athena R Brunt
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia
| | - Mark R Forwood
- School of Medical Science, Griffith University, Gold Coast, Queensland, Australia
| | | | - Mark J Tobin
- Australian Synchrotron, Clayton, Victoria, Australia
| | - T John Martin
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; The University of Melbourne, Department of Medicine at St. Vincent's Hospital, Fitzroy, Victoria, Australia
| | - Natalie A Sims
- St. Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; The University of Melbourne, Department of Medicine at St. Vincent's Hospital, Fitzroy, Victoria, Australia.
| |
Collapse
|
55
|
Idolazzi L, Rossini M, Viapiana O, Braga V, Fassio A, Benini C, Kunnathully V, Adami S, Gatti D. Teriparatide and denosumab combination therapy and skeletal metabolism. Osteoporos Int 2016; 27:3301-3307. [PMID: 27250971 DOI: 10.1007/s00198-016-3647-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Accepted: 05/19/2016] [Indexed: 01/22/2023]
Abstract
UNLABELLED Several therapies are available for osteoporis. Understanding the bone turnover changes and their mutual realtionship gives an overall view and might lead to a target therapy INTRODUCTION: The aim of this study is to compare the changes in bone turnover markers in patients treated with either denosumab alone, teriparatide (TPTD) alone, or in a third therapeutic scheme, when TPTD was added to patients previously treated with denosumab. METHODS Fifty-nine women over 65 years old with severe postmenopausal osteoporosis (evidence of at least two moderate-severe vertebral fractures) were enrolled in the study. Serum samples were collected every 3 months. They were assayed for intact N-propeptide of type I collagen (P1NP), C-terminal telopeptide of type I collagen (CTX), intact parathyroid hormone (PTH), 25 hydroxy-vitamin D (25 OHD), Sclerostin (SOST), and Dickkopf-related protein 1 (DKK1). Bone mass density was assessed by dual-energy X-ray absorptiometry at the lumbar spine and at the total hip. RESULTS In the groups treated only with TPTD or with denosumab, bone turnover markers increased and decreased, respectively. In TPTD group, a later significant increase in DKK1 was observed, while in denosumab group, a progressive increase in SOST was associated with a progressive significant decrease in DKK1. In the group treated first with denosumab and in which TPTD was added 3 months later, both CTX and P1NP increased 3 months after the beginning of TPTD. The strong effect of denosumab on bone turnover seems to be reversed by TPTD treatment. CONCLUSIONS In this study, we showed that TPTD is able to express its biological activity even when bone turnover is fully suppressed by denosumab treatment. The combination therapy is associated with significant increases in both DKK1 and SOST.
Collapse
Affiliation(s)
- L Idolazzi
- Rheumatology Unit - Department of Medicine, University of Verona, Piazzale A. Stefani 1, 37126, Verona, VR, Italy.
| | - M Rossini
- Rheumatology Unit - Department of Medicine, University of Verona, Piazzale A. Stefani 1, 37126, Verona, VR, Italy
| | - O Viapiana
- Rheumatology Unit - Department of Medicine, University of Verona, Piazzale A. Stefani 1, 37126, Verona, VR, Italy
| | - V Braga
- Rheumatology Unit - Department of Medicine, University of Verona, Piazzale A. Stefani 1, 37126, Verona, VR, Italy
| | - A Fassio
- Rheumatology Unit - Department of Medicine, University of Verona, Piazzale A. Stefani 1, 37126, Verona, VR, Italy
| | - C Benini
- Rheumatology Unit - Department of Medicine, University of Verona, Piazzale A. Stefani 1, 37126, Verona, VR, Italy
| | - V Kunnathully
- Rheumatology Unit - Department of Medicine, University of Verona, Piazzale A. Stefani 1, 37126, Verona, VR, Italy
| | - S Adami
- Rheumatology Unit - Department of Medicine, University of Verona, Piazzale A. Stefani 1, 37126, Verona, VR, Italy
| | - D Gatti
- Rheumatology Unit - Department of Medicine, University of Verona, Piazzale A. Stefani 1, 37126, Verona, VR, Italy
| |
Collapse
|
56
|
Hikata T, Hasegawa T, Horiuchi K, Fujita N, Iwanami A, Watanabe K, Ishii K, Nakamura M, Amizuka N, Matsumoto M. Histomorphometric analysis of minimodeling in the vertebrae in postmenopausal patients treated with anti-osteoporotic agents. Bone Rep 2016; 5:286-291. [PMID: 28580398 PMCID: PMC5440780 DOI: 10.1016/j.bonr.2016.10.002] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 10/06/2016] [Accepted: 10/12/2016] [Indexed: 02/03/2023] Open
Abstract
Minimodeling is a type of focal bone formation that is characterized by the lack of precedent bone erosion by osteoclasts. Although this form of bone formation has been described for more than a decade, how anti-osteoporotic agents that are currently used in clinical practice affect the kinetics of minimodeling is not fully understood. We performed a bone morphometric analysis using human vertebral specimens collected from postmenopausal patients who underwent spinal surgery. Patients were divided into three groups according to osteoporosis medication; non-treated, Eldecalcitol (ELD, a vitamin D derivative that has recently been approved to treat patients with osteoporosis in Japan)-treated, and bisphosphonate-treated groups. Five to six patients were enrolled in each group. There was a trend toward enhanced minimodeling in ELD-treated patients and suppressed of it in bisphosphonate-treated patients compared with untreated patients. The differences of minimodeling activity between ELD-treated and bisphosphonate-treated patients were statistically significant. The present study suggests that ELD and bisphosphonates have opposite effects on minimodeling from one another, and show that minimodeling also takes place in vertebrae as has been described for the ilium and femoral head in humans. We aimed to evaluate the potential effects of a new vitamin D3 analog, Eldecalcitol and BPs on minimodeling in humans using surgical specimens. We found that minimodeling is readily observed in the humans. We found that patients who had been treated with Eldecalcitol prior to surgery tend to have increased minimodeling activity. Our data indicate that Eldecalcitol can activate minimodeling in patients with post-menopausal osteoporosis while bisphosphonates have negative or little impact on this form of bone formation.
Collapse
Affiliation(s)
- Tomohiro Hikata
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan.,Department of Orthopedic Surgery, Kitasato Institute Hospital, Tokyo 108-8642, Japan
| | - Tomoka Hasegawa
- Department of Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Hokkaido University, Hokkaido 060-8586, Japan
| | - Keisuke Horiuchi
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Nobuyuki Fujita
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Akio Iwanami
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Kota Watanabe
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Ken Ishii
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Masaya Nakamura
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Norio Amizuka
- Department of Developmental Biology of Hard Tissue, Graduate School of Dental Medicine, Hokkaido University, Hokkaido 060-8586, Japan
| | - Morio Matsumoto
- Department of Orthopedic Surgery, Keio University School of Medicine, Tokyo 160-8582, Japan
| |
Collapse
|
57
|
Niimi R, Kono T, Nishihara A, Hasegawa M, Kono T, Sudo A. The Relationship of BMD Increases Between the First 12 Months and the Latter 12 Months by Daily Teriparatide Treatment. Calcif Tissue Int 2016; 99:282-8. [PMID: 27206528 DOI: 10.1007/s00223-016-0151-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 05/09/2016] [Indexed: 10/21/2022]
Abstract
The degree of correlation between the first 12 months and the latter 12 months of increased bone mineral density (BMD) with teriparatide treatment is unknown. We retrospectively investigated the correlation between the first 12 months and the latter 12 months of increased BMD owing to teriparatide treatment. We retrospectively analyzed 357 patients (mean age, 78 years) with osteoporosis treated with teriparatide 20 μg/day for 24 months. The primary efficacy measure was the correlation between lumbar spine (LS) and femoral neck (FN) BMD increases from baseline to 12 months and from 12 to 24 months. The correlation between the first 12 months and the latter 12 months of increased BMD was evaluated. We investigated the correlation between the increases in BMD and the baseline procollagen type I N-terminal propeptide (PINP) concentration. LS BMD significantly increased by 9.7 ± 8.3 % in the first 12 months and 3.5 ± 4.8 % in the latter 12 months. FN BMD increased by 2.2 ± 8.4 % in the first 12 months and 1.3 ± 4.9 % in the latter 12 months. Increased LS and FN BMD were not significantly correlated between the first 12 months and the latter 12 months. The serum baseline PINP concentration was correlated with the LS BMD in the first 12 months, and similarly, the PINP concentration at 12 months was correlated with the latter 12 months of increased LS BMD. Increased BMD by teriparatide treatment in the first 12 months and the latter 12 months was not significantly correlated.
Collapse
Affiliation(s)
- Rui Niimi
- Department of Orthopaedic Surgery, Tomidahama Hospital, 26-14 Tomidahamacho, Yokkaichi, Mie, 510-8008, Japan.
| | - Toshibumi Kono
- Department of Orthopaedic Surgery, Tomidahama Hospital, 26-14 Tomidahamacho, Yokkaichi, Mie, 510-8008, Japan
| | - Atsushi Nishihara
- Department of Orthopaedic Surgery, Tomidahama Hospital, 26-14 Tomidahamacho, Yokkaichi, Mie, 510-8008, Japan
| | - Masahiro Hasegawa
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| | - Toshihiko Kono
- Department of Orthopaedic Surgery, Tomidahama Hospital, 26-14 Tomidahamacho, Yokkaichi, Mie, 510-8008, Japan
| | - Akihiro Sudo
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, Tsu, Japan
| |
Collapse
|
58
|
Cavani F, Ferretti M, Smargiassi A, Palumbo C. PTH(1-34) effects on repairing experimentally drilled holes in rat femur: novel aspects - qualitative vs. quantitative improvement of osteogenesis. J Anat 2016; 230:75-84. [PMID: 27523886 DOI: 10.1111/joa.12533] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2016] [Indexed: 12/26/2022] Open
Abstract
The timetable of effects on bone repair of the active fraction-parathyroid hormone, PTH(1-34), was analytically investigated from the morphometric viewpoint in 3-month-old male Sprague-Dawley rats, whose femurs were drilled at mid-diaphyseal level (transcortical holes). The animals were divided into groups with/without PTH(1-34) administration, and sacrificed at different times (10, 28, 45 days after surgery). The observations reported here need to be framed in the context of our previous investigations regarding bone histogenesis (Ferretti et al. Anat Embryol. 2002; 206: 21-29) in which we demonstrated the occurrence of two successive bone-forming processes during both skeletal organogenesis and bone repair, i.e. static and dynamic osteogenesis: the former (due to stationary osteoblasts, haphazardly grouped in cords) producing preliminary bad quality trabecular bone, the latter (due to typical polarized osteoblasts organized in ordered movable laminae) producing mechanically valid bone tissue. The primary function of static osteogenesis is to provide a rigid scaffold containing osteocytes (i.e. mechano-sensors) for osteoblast laminae acting in dynamic osteogenesis. In the present work, histomorphometric analysis revealed that, already 10 days after drilling, despite the holes being temporarily filled by the same amount of newly formed trabecular bone by static osteogenesis independently of the treatment, the extent of the surface of movable osteoblast-laminae (covering the trabecular surface) was statistically higher in animals submitted to PTH(1-34) administration than in control ones; this datum strongly suggests the effect of PTH(1-34) alone in anticipating the occurrence of dynamic osteogenesis involved in the production of good quality bone (with more ordered collagen texture) more suitable for loading. This study could be crucial in further translational clinical research in humans for defining the best therapeutic strategies to be applied in recovering severe skeletal lesions, particularly as regards the time of PTH(1-34) administration.
Collapse
Affiliation(s)
- Francesco Cavani
- Dipartimento di Scienze Biomediche, Metaboliche e Neuroscienze - Sez. Morfologia umana, Università di Modena e Reggio Emilia, Modena, Italy
| | - Marzia Ferretti
- Dipartimento di Scienze Biomediche, Metaboliche e Neuroscienze - Sez. Morfologia umana, Università di Modena e Reggio Emilia, Modena, Italy
| | - Alberto Smargiassi
- Dipartimento di Scienze Biomediche, Metaboliche e Neuroscienze - Sez. Morfologia umana, Università di Modena e Reggio Emilia, Modena, Italy
| | - Carla Palumbo
- Dipartimento di Scienze Biomediche, Metaboliche e Neuroscienze - Sez. Morfologia umana, Università di Modena e Reggio Emilia, Modena, Italy
| |
Collapse
|
59
|
Lindsay R, Krege JH, Marin F, Jin L, Stepan JJ. Teriparatide for osteoporosis: importance of the full course. Osteoporos Int 2016; 27:2395-410. [PMID: 26902094 PMCID: PMC4947115 DOI: 10.1007/s00198-016-3534-6] [Citation(s) in RCA: 118] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/09/2016] [Indexed: 11/01/2022]
Abstract
Teriparatide (TPTD) is the only currently available therapeutic agent that increases the formation of new bone tissue and can provide some remediation of the architectural defects in the osteoporotic skeleton. The use of teriparatide clinically is limited to 24 months. We review clinical findings during daily teriparatide treatment over time. Teriparatide appears to increase bone formation more than bone resorption as determined biochemically and histologically. Teriparatide exerts its positive effects on bone formation in two distinct fashions. The first is direct stimulation of bone formation that occurs within active remodeling sites (remodeling-based bone formation) and on surfaces of bone previously inactive (modeling-based bone formation). The second is an increase in the initiation of new remodeling sites. Both processes contribute to the final increase in bone density observed by non-invasive tools such as DXA. Remodeling is the repair process by which skeletal tissue is maintained in a young healthy state, and when stimulated by TPTD is associated with a positive bone balance within each remodeling cavity. It seems likely therefore that this component will contribute to the anti-fracture efficacy of TPTD. Teriparatide reduces the risk of fracture, and this effect appears to increase with longer duration of therapy. The use of novel treatment regimens, including shorter courses, should be held in abeyance until controlled clinical trials are completed to define the relative fracture benefits of such approaches in comparison to the 24-month daily use of the agent. Summary In patients with osteoporosis at high risk for fracture, the full continuous 24-month course with teriparatide results in improved skeletal health and outcomes than shorter time periods.
Collapse
Affiliation(s)
- R Lindsay
- Helen Hayes Hospital, West Haverstraw, NY, USA
| | - J H Krege
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA.
| | - F Marin
- Lilly Research Centre, Eli Lilly and Company, Windlesham, Surrey, UK
| | - L Jin
- Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, USA
| | - J J Stepan
- Institute of Rheumatology, Faculty of Medicine 1, Charles University, Prague, Czech Republic
| |
Collapse
|
60
|
Martin TJ. Parathyroid Hormone-Related Protein, Its Regulation of Cartilage and Bone Development, and Role in Treating Bone Diseases. Physiol Rev 2016; 96:831-71. [DOI: 10.1152/physrev.00031.2015] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Although parathyroid hormone-related protein (PTHrP) was discovered as a cancer-derived hormone, it has been revealed as an important paracrine/autocrine regulator in many tissues, where its effects are context dependent. Thus its location and action in the vasculature explained decades-long observations that injection of PTH into animals rapidly lowered blood pressure by producing vasodilatation. Its roles have been specified in development and maturity in cartilage and bone as a crucial regulator of endochondral bone formation and bone remodeling, respectively. Although it shares actions with parathyroid hormone (PTH) through the use of their common receptor, PTHR1, PTHrP has other actions mediated by regions within the molecule beyond the amino-terminal sequence that resembles PTH, including the ability to promote placental transfer of calcium from mother to fetus. A striking feature of the physiology of PTHrP is that it possesses structural features that equip it to be transported in and out of the nucleus, and makes use of a specific nuclear import mechanism to do so. Evidence from mouse genetic experiments shows that PTHrP generated locally in bone is essential for normal bone remodeling. Whereas the main physiological function of PTH is the hormonal regulation of calcium metabolism, locally generated PTHrP is the important physiological mediator of bone remodeling postnatally. Thus the use of intermittent injection of PTH as an anabolic therapy for bone appears to be a pharmacological application of the physiological function of PTHrP. There is much current interest in the possibility of developing PTHrP analogs that might enhance the therapeutic anabolic effects.
Collapse
Affiliation(s)
- T. John Martin
- St Vincent's Institute of Medical Research, Department of Medicine, University of Melbourne, St Vincent's Hospital, Melbourne, Australia
| |
Collapse
|
61
|
Yamamoto T, Hasegawa T, Sasaki M, Hongo H, Tsuboi K, Shimizu T, Ota M, Haraguchi M, Takahata M, Oda K, Luiz de Freitas PH, Takakura A, Takao-Kawabata R, Isogai Y, Amizuka N. Frequency of Teriparatide Administration Affects the Histological Pattern of Bone Formation in Young Adult Male Mice. Endocrinology 2016; 157:2604-20. [PMID: 27227535 DOI: 10.1210/en.2015-2028] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Evidence supports that daily and once-weekly administration of teriparatide, human (h)PTH(1-34), enhance bone mass in osteoporotic patients. However, it is uncertain whether different frequencies of hPTH(1-34) administration would induce bone formation similarly in terms of quantity and quality. To investigate that issue, mice were subjected to different frequencies of PTH administration, and their bones were histologically examined. Frequencies of administration were 1 time/2 days, 1 time a day, and 2 and 4 times a day. Mice were allocated to either to control or to 3 different dosing regimens: 80 μg/kg of hPTH(1-34) per injection (80 μg/kg per dose), 80 μg/kg of hPTH(1-34) per day (80 μg/kg · d), or 20 μg/kg of hPTH(1-34) per day (20 μg/kg · d). With the regimens of 80 μg/kg per dose and 80 μg/kg · d, high-frequency hPTH(1-34) administration increased metaphyseal trabecular number. However, 4 doses per day induced the formation of thin trabeculae, whereas the daily PTH regimen resulted in thicker trabeculae. A similar pattern was observed with the lower daily hPTH(1-34) dose (20 μg/kg · d): more frequent PTH administration led to the formation of thin trabeculae, showing a thick preosteoblastic cell layer, several osteoclasts, and scalloped cement lines that indicated accelerated bone remodeling. On the other hand, low-frequency PTH administration induced new bone with mature osteoblasts lying on mildly convex surfaces representative of arrest lines, which suggests minimodeling-based bone formation. Thus, high-frequency PTH administration seems to increase bone mass rapidly by forming thin trabeculae through accelerated bone remodeling. Alternatively, low-frequency PTH administration leads to the formation of thicker trabeculae through bone remodeling and minimodeling.
Collapse
Affiliation(s)
- Tomomaya Yamamoto
- Department of Developmental Biology of Hard Tissue (T.Y., T.H., H.H., K.T., M.H., N.A.), Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586 Japan; Department of Applied Prosthodontics (M.S.), Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588 Japan; Department of Orthopedic Surgery Graduate School of Medicine (T.S., M.O., M.T.), Hokkaido University, Sapporo, 951-8514 Japan; Division of Biochemistry (K.O.), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514 Japan; Department of Dentistry (P.H.L.d.F.), Federal University of Sergipe at Lagarto, Campus Legarto, 49400-000 Brazil; and Asahi Kasei Pharma Co. Ltd (A.T., R.T.-K., Y.I.), Tokyo, 101-8101 Japan
| | - Tomoka Hasegawa
- Department of Developmental Biology of Hard Tissue (T.Y., T.H., H.H., K.T., M.H., N.A.), Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586 Japan; Department of Applied Prosthodontics (M.S.), Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588 Japan; Department of Orthopedic Surgery Graduate School of Medicine (T.S., M.O., M.T.), Hokkaido University, Sapporo, 951-8514 Japan; Division of Biochemistry (K.O.), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514 Japan; Department of Dentistry (P.H.L.d.F.), Federal University of Sergipe at Lagarto, Campus Legarto, 49400-000 Brazil; and Asahi Kasei Pharma Co. Ltd (A.T., R.T.-K., Y.I.), Tokyo, 101-8101 Japan
| | - Muneteru Sasaki
- Department of Developmental Biology of Hard Tissue (T.Y., T.H., H.H., K.T., M.H., N.A.), Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586 Japan; Department of Applied Prosthodontics (M.S.), Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588 Japan; Department of Orthopedic Surgery Graduate School of Medicine (T.S., M.O., M.T.), Hokkaido University, Sapporo, 951-8514 Japan; Division of Biochemistry (K.O.), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514 Japan; Department of Dentistry (P.H.L.d.F.), Federal University of Sergipe at Lagarto, Campus Legarto, 49400-000 Brazil; and Asahi Kasei Pharma Co. Ltd (A.T., R.T.-K., Y.I.), Tokyo, 101-8101 Japan
| | - Hiromi Hongo
- Department of Developmental Biology of Hard Tissue (T.Y., T.H., H.H., K.T., M.H., N.A.), Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586 Japan; Department of Applied Prosthodontics (M.S.), Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588 Japan; Department of Orthopedic Surgery Graduate School of Medicine (T.S., M.O., M.T.), Hokkaido University, Sapporo, 951-8514 Japan; Division of Biochemistry (K.O.), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514 Japan; Department of Dentistry (P.H.L.d.F.), Federal University of Sergipe at Lagarto, Campus Legarto, 49400-000 Brazil; and Asahi Kasei Pharma Co. Ltd (A.T., R.T.-K., Y.I.), Tokyo, 101-8101 Japan
| | - Kanako Tsuboi
- Department of Developmental Biology of Hard Tissue (T.Y., T.H., H.H., K.T., M.H., N.A.), Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586 Japan; Department of Applied Prosthodontics (M.S.), Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588 Japan; Department of Orthopedic Surgery Graduate School of Medicine (T.S., M.O., M.T.), Hokkaido University, Sapporo, 951-8514 Japan; Division of Biochemistry (K.O.), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514 Japan; Department of Dentistry (P.H.L.d.F.), Federal University of Sergipe at Lagarto, Campus Legarto, 49400-000 Brazil; and Asahi Kasei Pharma Co. Ltd (A.T., R.T.-K., Y.I.), Tokyo, 101-8101 Japan
| | - Tomohiro Shimizu
- Department of Developmental Biology of Hard Tissue (T.Y., T.H., H.H., K.T., M.H., N.A.), Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586 Japan; Department of Applied Prosthodontics (M.S.), Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588 Japan; Department of Orthopedic Surgery Graduate School of Medicine (T.S., M.O., M.T.), Hokkaido University, Sapporo, 951-8514 Japan; Division of Biochemistry (K.O.), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514 Japan; Department of Dentistry (P.H.L.d.F.), Federal University of Sergipe at Lagarto, Campus Legarto, 49400-000 Brazil; and Asahi Kasei Pharma Co. Ltd (A.T., R.T.-K., Y.I.), Tokyo, 101-8101 Japan
| | - Masahiro Ota
- Department of Developmental Biology of Hard Tissue (T.Y., T.H., H.H., K.T., M.H., N.A.), Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586 Japan; Department of Applied Prosthodontics (M.S.), Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588 Japan; Department of Orthopedic Surgery Graduate School of Medicine (T.S., M.O., M.T.), Hokkaido University, Sapporo, 951-8514 Japan; Division of Biochemistry (K.O.), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514 Japan; Department of Dentistry (P.H.L.d.F.), Federal University of Sergipe at Lagarto, Campus Legarto, 49400-000 Brazil; and Asahi Kasei Pharma Co. Ltd (A.T., R.T.-K., Y.I.), Tokyo, 101-8101 Japan
| | - Mai Haraguchi
- Department of Developmental Biology of Hard Tissue (T.Y., T.H., H.H., K.T., M.H., N.A.), Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586 Japan; Department of Applied Prosthodontics (M.S.), Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588 Japan; Department of Orthopedic Surgery Graduate School of Medicine (T.S., M.O., M.T.), Hokkaido University, Sapporo, 951-8514 Japan; Division of Biochemistry (K.O.), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514 Japan; Department of Dentistry (P.H.L.d.F.), Federal University of Sergipe at Lagarto, Campus Legarto, 49400-000 Brazil; and Asahi Kasei Pharma Co. Ltd (A.T., R.T.-K., Y.I.), Tokyo, 101-8101 Japan
| | - Masahiko Takahata
- Department of Developmental Biology of Hard Tissue (T.Y., T.H., H.H., K.T., M.H., N.A.), Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586 Japan; Department of Applied Prosthodontics (M.S.), Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588 Japan; Department of Orthopedic Surgery Graduate School of Medicine (T.S., M.O., M.T.), Hokkaido University, Sapporo, 951-8514 Japan; Division of Biochemistry (K.O.), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514 Japan; Department of Dentistry (P.H.L.d.F.), Federal University of Sergipe at Lagarto, Campus Legarto, 49400-000 Brazil; and Asahi Kasei Pharma Co. Ltd (A.T., R.T.-K., Y.I.), Tokyo, 101-8101 Japan
| | - Kimimitsu Oda
- Department of Developmental Biology of Hard Tissue (T.Y., T.H., H.H., K.T., M.H., N.A.), Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586 Japan; Department of Applied Prosthodontics (M.S.), Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588 Japan; Department of Orthopedic Surgery Graduate School of Medicine (T.S., M.O., M.T.), Hokkaido University, Sapporo, 951-8514 Japan; Division of Biochemistry (K.O.), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514 Japan; Department of Dentistry (P.H.L.d.F.), Federal University of Sergipe at Lagarto, Campus Legarto, 49400-000 Brazil; and Asahi Kasei Pharma Co. Ltd (A.T., R.T.-K., Y.I.), Tokyo, 101-8101 Japan
| | - Paulo Henrique Luiz de Freitas
- Department of Developmental Biology of Hard Tissue (T.Y., T.H., H.H., K.T., M.H., N.A.), Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586 Japan; Department of Applied Prosthodontics (M.S.), Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588 Japan; Department of Orthopedic Surgery Graduate School of Medicine (T.S., M.O., M.T.), Hokkaido University, Sapporo, 951-8514 Japan; Division of Biochemistry (K.O.), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514 Japan; Department of Dentistry (P.H.L.d.F.), Federal University of Sergipe at Lagarto, Campus Legarto, 49400-000 Brazil; and Asahi Kasei Pharma Co. Ltd (A.T., R.T.-K., Y.I.), Tokyo, 101-8101 Japan
| | - Aya Takakura
- Department of Developmental Biology of Hard Tissue (T.Y., T.H., H.H., K.T., M.H., N.A.), Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586 Japan; Department of Applied Prosthodontics (M.S.), Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588 Japan; Department of Orthopedic Surgery Graduate School of Medicine (T.S., M.O., M.T.), Hokkaido University, Sapporo, 951-8514 Japan; Division of Biochemistry (K.O.), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514 Japan; Department of Dentistry (P.H.L.d.F.), Federal University of Sergipe at Lagarto, Campus Legarto, 49400-000 Brazil; and Asahi Kasei Pharma Co. Ltd (A.T., R.T.-K., Y.I.), Tokyo, 101-8101 Japan
| | - Ryoko Takao-Kawabata
- Department of Developmental Biology of Hard Tissue (T.Y., T.H., H.H., K.T., M.H., N.A.), Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586 Japan; Department of Applied Prosthodontics (M.S.), Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588 Japan; Department of Orthopedic Surgery Graduate School of Medicine (T.S., M.O., M.T.), Hokkaido University, Sapporo, 951-8514 Japan; Division of Biochemistry (K.O.), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514 Japan; Department of Dentistry (P.H.L.d.F.), Federal University of Sergipe at Lagarto, Campus Legarto, 49400-000 Brazil; and Asahi Kasei Pharma Co. Ltd (A.T., R.T.-K., Y.I.), Tokyo, 101-8101 Japan
| | - Yukihiro Isogai
- Department of Developmental Biology of Hard Tissue (T.Y., T.H., H.H., K.T., M.H., N.A.), Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586 Japan; Department of Applied Prosthodontics (M.S.), Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588 Japan; Department of Orthopedic Surgery Graduate School of Medicine (T.S., M.O., M.T.), Hokkaido University, Sapporo, 951-8514 Japan; Division of Biochemistry (K.O.), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514 Japan; Department of Dentistry (P.H.L.d.F.), Federal University of Sergipe at Lagarto, Campus Legarto, 49400-000 Brazil; and Asahi Kasei Pharma Co. Ltd (A.T., R.T.-K., Y.I.), Tokyo, 101-8101 Japan
| | - Norio Amizuka
- Department of Developmental Biology of Hard Tissue (T.Y., T.H., H.H., K.T., M.H., N.A.), Graduate School of Dental Medicine, Hokkaido University, Sapporo, 060-8586 Japan; Department of Applied Prosthodontics (M.S.), Unit of Translational Medicine, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, 852-8588 Japan; Department of Orthopedic Surgery Graduate School of Medicine (T.S., M.O., M.T.), Hokkaido University, Sapporo, 951-8514 Japan; Division of Biochemistry (K.O.), Graduate School of Medical and Dental Sciences, Niigata University, Niigata, 951-8514 Japan; Department of Dentistry (P.H.L.d.F.), Federal University of Sergipe at Lagarto, Campus Legarto, 49400-000 Brazil; and Asahi Kasei Pharma Co. Ltd (A.T., R.T.-K., Y.I.), Tokyo, 101-8101 Japan
| |
Collapse
|
62
|
Oestrogen and parathyroid hormone alleviate lumbar intervertebral disc degeneration in ovariectomized rats and enhance Wnt/β-catenin pathway activity. Sci Rep 2016; 6:27521. [PMID: 27279629 PMCID: PMC4899752 DOI: 10.1038/srep27521] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 05/09/2016] [Indexed: 11/08/2022] Open
Abstract
To investigate the mitigation effect and mechanism of oestrogen and PTH on disc degeneration in rats after ovariectomy, as well as on Wnt/β-catenin pathway activity, thirty 3-month-old rats were ovariectomized and divided into three groups. Ten additional rats were used as controls. Eight weeks later, the rats were administered oestrogen or PTH for 12 weeks, and then discs were collected for tests. Results showed that nucleus pulposus cells in the Sham group were mostly notochord cells, while in the OVX group, cells gradually developed into chondrocyte-like cells. Oestrogen or PTH could partly recover the notochord cell number. After ovariectomy, the endplate roughened and endplate porosity decreased. After oestrogen or PTH treatment, the smoothness and porosity of endplate recovered. Compared with the Sham group, Aggrecan, Col2a and Wnt/β-catenin pathway expression in OVX group decreased, and either oestrogen or PTH treatment improved their expression. The biomechanical properties of intervertebral disc significantly changed after ovariectomy, and oestrogen or PTH treatment partly recovered them. Disc degeneration occurred with low oestrogen, and the underlying mechanisms involve nutrition supply disorders, cell type changes and decreased Wnt/β-catenin pathway activity. Oestrogen and PTH can retard disc degeneration in OVX rats and enhance Wnt/β-catenin pathway activity in nucleus pulposus.
Collapse
|
63
|
Eriksen EF, Brown JP. Commentary: Concurrent administration of PTH and antiresorptives: Additive effects or DXA cosmetics. Bone 2016; 86:139-42. [PMID: 26944033 DOI: 10.1016/j.bone.2016.02.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Revised: 02/15/2016] [Accepted: 02/20/2016] [Indexed: 01/08/2023]
Abstract
Osteoanabolic therapy with parathyroid hormone (PTH(1-84)) or the PTH analogues teriparatide (PTH(1-34), TPTD) and abaloparatide induces a positive remodeling balance and increases modeling and remodeling activity on bone surfaces. As the anabolic action of PTH is primarily remodeling based increased bone turnover maximizes bone accrual. Increased remodeling, however, also increases cortical porosity and reduces mineralization of newly formed bone, which may cause initial reductions in BMD, particularly at sites rich in cortical bone. Increased cortical porosity may also have negative consequences for bone strength. Consequently, an interest developed in concurrent therapies offsetting the potential early negative cortical bone effects developed, and several studies using varying concurrent combinations of TPTD or PTH(1-84) with various antiresorptive (anti-catabolic) agents (estrogen, SERMs, bisphosphonates and denosumab) have been published. This commentary addresses the discrepancy between changes in areal bone mineral density (BMD) and bone turnover markers (BTM) in concurrent therapy studies leading to possible misinterpretations of the results. In studies of concurrent therapies increases in BMD are generally accompanied by decreases in biochemical markers of bone turnover. This includes Procollagen Type I N-Terminal Propetide (PINP), which has emerged as a reliable marker of bone formation during osteoanabolic therapy. We therefore want to submit, that the larger increases in BMD seen initially in patients on concurrent therapy mask the potential for later reduced osteoanabolic action of PTH. This notion is corroborated by: 1) the lesser impairment of bone anabolism seen with milder antiresorptive modalities like hormone replacement therapy (HRT) or Selective Estrogen Receptor Modulators (SERMs); 2) the changes in BMD seen in extension studies where treatment naïve patients previously treated with PTH alone are crossed over to antiresorptive drugs. We therefore advise against a general use of concurrent therapy with PTH and antiresorptive agents, as it entails blunting of osteoanabolic action of PTH in the long run.
Collapse
Affiliation(s)
- Erik Fink Eriksen
- Dept. of Clinical Endocrinology, Morbid Obesity and Preventive Medicine, Oslo University Hospital, Norway
| | - Jacques P Brown
- Dept. of Rheumatology, Laval University and CHU de Québec (CHUL) Research Centre, Quebec City, Canada
| |
Collapse
|
64
|
Dempster DW, Zhou H, Recker RR, Brown JP, Recknor CP, Lewiecki EM, Miller PD, Rao SD, Kendler DL, Lindsay R, Krege JH, Alam J, Taylor KA, Janos B, Ruff VA. Differential Effects of Teriparatide and Denosumab on Intact PTH and Bone Formation Indices: AVA Osteoporosis Study. J Clin Endocrinol Metab 2016; 101:1353-63. [PMID: 26859106 PMCID: PMC4880160 DOI: 10.1210/jc.2015-4181] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We compared effects of teriparatide and denosumab on PTH, bone turnover markers, and bone histomorphometry in osteoporotic postmenopausal women. The findings were inconsistent with an early indirect anabolic effect of denosumab.
Collapse
Affiliation(s)
- David W Dempster
- Regional Bone Center (D.W.D., H.Z., R.L.), Helen Hayes Hospital, West Haverstraw, New York; Department of Pathology and Cell Biology (D.W.D.), College of Physicians and Surgeons of Columbia University, New York; Department of Medicine (R.R.R.), Division of Endocrinology, School of Medicine, Creighton University, Omaha, Nebraska; Rheumatology and Bone Diseases Research Group (J.P.B.), CHU de Quebec (CHUL) Research Centre and Department of Medicine, Laval University, Quebec City, Quebec, Canada; United Osteoporosis Centers (C.P.R.), Gainesville, Georgia; New Mexico Clinical Research & Osteoporosis Center (E.M.L.), Albuquerque, New Mexico; Department of Medicine (P.D.M.), Colorado Center for Bone Research, Lakewood, Colorado; Bone & Mineral Research Laboratory (S.D.R.), Henry Ford Hospital, Detroit, Michigan; Department of Medicine (Endocrinology) (D.L.K.), University of British Columbia, Vancouver, British Columbia, Canada; Department of Medicine (R.L.), College of Physicians and Surgeons of Columbia University, New York; Lilly Research Laboratories (J.H.K., J.A.), Eli Lilly and Company, Indianapolis, Indiana; Musculoskeletal and Men's Health (K.A.T., V.A.R.), Lilly USA, LLC, Indianapolis, Indiana; Research and Development - Bio-Medicines (B.J.), Eli Lilly Canada Inc., Toronto, Ontario, Canada
| | - Hua Zhou
- Regional Bone Center (D.W.D., H.Z., R.L.), Helen Hayes Hospital, West Haverstraw, New York; Department of Pathology and Cell Biology (D.W.D.), College of Physicians and Surgeons of Columbia University, New York; Department of Medicine (R.R.R.), Division of Endocrinology, School of Medicine, Creighton University, Omaha, Nebraska; Rheumatology and Bone Diseases Research Group (J.P.B.), CHU de Quebec (CHUL) Research Centre and Department of Medicine, Laval University, Quebec City, Quebec, Canada; United Osteoporosis Centers (C.P.R.), Gainesville, Georgia; New Mexico Clinical Research & Osteoporosis Center (E.M.L.), Albuquerque, New Mexico; Department of Medicine (P.D.M.), Colorado Center for Bone Research, Lakewood, Colorado; Bone & Mineral Research Laboratory (S.D.R.), Henry Ford Hospital, Detroit, Michigan; Department of Medicine (Endocrinology) (D.L.K.), University of British Columbia, Vancouver, British Columbia, Canada; Department of Medicine (R.L.), College of Physicians and Surgeons of Columbia University, New York; Lilly Research Laboratories (J.H.K., J.A.), Eli Lilly and Company, Indianapolis, Indiana; Musculoskeletal and Men's Health (K.A.T., V.A.R.), Lilly USA, LLC, Indianapolis, Indiana; Research and Development - Bio-Medicines (B.J.), Eli Lilly Canada Inc., Toronto, Ontario, Canada
| | - Robert R Recker
- Regional Bone Center (D.W.D., H.Z., R.L.), Helen Hayes Hospital, West Haverstraw, New York; Department of Pathology and Cell Biology (D.W.D.), College of Physicians and Surgeons of Columbia University, New York; Department of Medicine (R.R.R.), Division of Endocrinology, School of Medicine, Creighton University, Omaha, Nebraska; Rheumatology and Bone Diseases Research Group (J.P.B.), CHU de Quebec (CHUL) Research Centre and Department of Medicine, Laval University, Quebec City, Quebec, Canada; United Osteoporosis Centers (C.P.R.), Gainesville, Georgia; New Mexico Clinical Research & Osteoporosis Center (E.M.L.), Albuquerque, New Mexico; Department of Medicine (P.D.M.), Colorado Center for Bone Research, Lakewood, Colorado; Bone & Mineral Research Laboratory (S.D.R.), Henry Ford Hospital, Detroit, Michigan; Department of Medicine (Endocrinology) (D.L.K.), University of British Columbia, Vancouver, British Columbia, Canada; Department of Medicine (R.L.), College of Physicians and Surgeons of Columbia University, New York; Lilly Research Laboratories (J.H.K., J.A.), Eli Lilly and Company, Indianapolis, Indiana; Musculoskeletal and Men's Health (K.A.T., V.A.R.), Lilly USA, LLC, Indianapolis, Indiana; Research and Development - Bio-Medicines (B.J.), Eli Lilly Canada Inc., Toronto, Ontario, Canada
| | - Jacques P Brown
- Regional Bone Center (D.W.D., H.Z., R.L.), Helen Hayes Hospital, West Haverstraw, New York; Department of Pathology and Cell Biology (D.W.D.), College of Physicians and Surgeons of Columbia University, New York; Department of Medicine (R.R.R.), Division of Endocrinology, School of Medicine, Creighton University, Omaha, Nebraska; Rheumatology and Bone Diseases Research Group (J.P.B.), CHU de Quebec (CHUL) Research Centre and Department of Medicine, Laval University, Quebec City, Quebec, Canada; United Osteoporosis Centers (C.P.R.), Gainesville, Georgia; New Mexico Clinical Research & Osteoporosis Center (E.M.L.), Albuquerque, New Mexico; Department of Medicine (P.D.M.), Colorado Center for Bone Research, Lakewood, Colorado; Bone & Mineral Research Laboratory (S.D.R.), Henry Ford Hospital, Detroit, Michigan; Department of Medicine (Endocrinology) (D.L.K.), University of British Columbia, Vancouver, British Columbia, Canada; Department of Medicine (R.L.), College of Physicians and Surgeons of Columbia University, New York; Lilly Research Laboratories (J.H.K., J.A.), Eli Lilly and Company, Indianapolis, Indiana; Musculoskeletal and Men's Health (K.A.T., V.A.R.), Lilly USA, LLC, Indianapolis, Indiana; Research and Development - Bio-Medicines (B.J.), Eli Lilly Canada Inc., Toronto, Ontario, Canada
| | - Christopher P Recknor
- Regional Bone Center (D.W.D., H.Z., R.L.), Helen Hayes Hospital, West Haverstraw, New York; Department of Pathology and Cell Biology (D.W.D.), College of Physicians and Surgeons of Columbia University, New York; Department of Medicine (R.R.R.), Division of Endocrinology, School of Medicine, Creighton University, Omaha, Nebraska; Rheumatology and Bone Diseases Research Group (J.P.B.), CHU de Quebec (CHUL) Research Centre and Department of Medicine, Laval University, Quebec City, Quebec, Canada; United Osteoporosis Centers (C.P.R.), Gainesville, Georgia; New Mexico Clinical Research & Osteoporosis Center (E.M.L.), Albuquerque, New Mexico; Department of Medicine (P.D.M.), Colorado Center for Bone Research, Lakewood, Colorado; Bone & Mineral Research Laboratory (S.D.R.), Henry Ford Hospital, Detroit, Michigan; Department of Medicine (Endocrinology) (D.L.K.), University of British Columbia, Vancouver, British Columbia, Canada; Department of Medicine (R.L.), College of Physicians and Surgeons of Columbia University, New York; Lilly Research Laboratories (J.H.K., J.A.), Eli Lilly and Company, Indianapolis, Indiana; Musculoskeletal and Men's Health (K.A.T., V.A.R.), Lilly USA, LLC, Indianapolis, Indiana; Research and Development - Bio-Medicines (B.J.), Eli Lilly Canada Inc., Toronto, Ontario, Canada
| | - E Michael Lewiecki
- Regional Bone Center (D.W.D., H.Z., R.L.), Helen Hayes Hospital, West Haverstraw, New York; Department of Pathology and Cell Biology (D.W.D.), College of Physicians and Surgeons of Columbia University, New York; Department of Medicine (R.R.R.), Division of Endocrinology, School of Medicine, Creighton University, Omaha, Nebraska; Rheumatology and Bone Diseases Research Group (J.P.B.), CHU de Quebec (CHUL) Research Centre and Department of Medicine, Laval University, Quebec City, Quebec, Canada; United Osteoporosis Centers (C.P.R.), Gainesville, Georgia; New Mexico Clinical Research & Osteoporosis Center (E.M.L.), Albuquerque, New Mexico; Department of Medicine (P.D.M.), Colorado Center for Bone Research, Lakewood, Colorado; Bone & Mineral Research Laboratory (S.D.R.), Henry Ford Hospital, Detroit, Michigan; Department of Medicine (Endocrinology) (D.L.K.), University of British Columbia, Vancouver, British Columbia, Canada; Department of Medicine (R.L.), College of Physicians and Surgeons of Columbia University, New York; Lilly Research Laboratories (J.H.K., J.A.), Eli Lilly and Company, Indianapolis, Indiana; Musculoskeletal and Men's Health (K.A.T., V.A.R.), Lilly USA, LLC, Indianapolis, Indiana; Research and Development - Bio-Medicines (B.J.), Eli Lilly Canada Inc., Toronto, Ontario, Canada
| | - Paul D Miller
- Regional Bone Center (D.W.D., H.Z., R.L.), Helen Hayes Hospital, West Haverstraw, New York; Department of Pathology and Cell Biology (D.W.D.), College of Physicians and Surgeons of Columbia University, New York; Department of Medicine (R.R.R.), Division of Endocrinology, School of Medicine, Creighton University, Omaha, Nebraska; Rheumatology and Bone Diseases Research Group (J.P.B.), CHU de Quebec (CHUL) Research Centre and Department of Medicine, Laval University, Quebec City, Quebec, Canada; United Osteoporosis Centers (C.P.R.), Gainesville, Georgia; New Mexico Clinical Research & Osteoporosis Center (E.M.L.), Albuquerque, New Mexico; Department of Medicine (P.D.M.), Colorado Center for Bone Research, Lakewood, Colorado; Bone & Mineral Research Laboratory (S.D.R.), Henry Ford Hospital, Detroit, Michigan; Department of Medicine (Endocrinology) (D.L.K.), University of British Columbia, Vancouver, British Columbia, Canada; Department of Medicine (R.L.), College of Physicians and Surgeons of Columbia University, New York; Lilly Research Laboratories (J.H.K., J.A.), Eli Lilly and Company, Indianapolis, Indiana; Musculoskeletal and Men's Health (K.A.T., V.A.R.), Lilly USA, LLC, Indianapolis, Indiana; Research and Development - Bio-Medicines (B.J.), Eli Lilly Canada Inc., Toronto, Ontario, Canada
| | - Sudhaker D Rao
- Regional Bone Center (D.W.D., H.Z., R.L.), Helen Hayes Hospital, West Haverstraw, New York; Department of Pathology and Cell Biology (D.W.D.), College of Physicians and Surgeons of Columbia University, New York; Department of Medicine (R.R.R.), Division of Endocrinology, School of Medicine, Creighton University, Omaha, Nebraska; Rheumatology and Bone Diseases Research Group (J.P.B.), CHU de Quebec (CHUL) Research Centre and Department of Medicine, Laval University, Quebec City, Quebec, Canada; United Osteoporosis Centers (C.P.R.), Gainesville, Georgia; New Mexico Clinical Research & Osteoporosis Center (E.M.L.), Albuquerque, New Mexico; Department of Medicine (P.D.M.), Colorado Center for Bone Research, Lakewood, Colorado; Bone & Mineral Research Laboratory (S.D.R.), Henry Ford Hospital, Detroit, Michigan; Department of Medicine (Endocrinology) (D.L.K.), University of British Columbia, Vancouver, British Columbia, Canada; Department of Medicine (R.L.), College of Physicians and Surgeons of Columbia University, New York; Lilly Research Laboratories (J.H.K., J.A.), Eli Lilly and Company, Indianapolis, Indiana; Musculoskeletal and Men's Health (K.A.T., V.A.R.), Lilly USA, LLC, Indianapolis, Indiana; Research and Development - Bio-Medicines (B.J.), Eli Lilly Canada Inc., Toronto, Ontario, Canada
| | - David L Kendler
- Regional Bone Center (D.W.D., H.Z., R.L.), Helen Hayes Hospital, West Haverstraw, New York; Department of Pathology and Cell Biology (D.W.D.), College of Physicians and Surgeons of Columbia University, New York; Department of Medicine (R.R.R.), Division of Endocrinology, School of Medicine, Creighton University, Omaha, Nebraska; Rheumatology and Bone Diseases Research Group (J.P.B.), CHU de Quebec (CHUL) Research Centre and Department of Medicine, Laval University, Quebec City, Quebec, Canada; United Osteoporosis Centers (C.P.R.), Gainesville, Georgia; New Mexico Clinical Research & Osteoporosis Center (E.M.L.), Albuquerque, New Mexico; Department of Medicine (P.D.M.), Colorado Center for Bone Research, Lakewood, Colorado; Bone & Mineral Research Laboratory (S.D.R.), Henry Ford Hospital, Detroit, Michigan; Department of Medicine (Endocrinology) (D.L.K.), University of British Columbia, Vancouver, British Columbia, Canada; Department of Medicine (R.L.), College of Physicians and Surgeons of Columbia University, New York; Lilly Research Laboratories (J.H.K., J.A.), Eli Lilly and Company, Indianapolis, Indiana; Musculoskeletal and Men's Health (K.A.T., V.A.R.), Lilly USA, LLC, Indianapolis, Indiana; Research and Development - Bio-Medicines (B.J.), Eli Lilly Canada Inc., Toronto, Ontario, Canada
| | - Robert Lindsay
- Regional Bone Center (D.W.D., H.Z., R.L.), Helen Hayes Hospital, West Haverstraw, New York; Department of Pathology and Cell Biology (D.W.D.), College of Physicians and Surgeons of Columbia University, New York; Department of Medicine (R.R.R.), Division of Endocrinology, School of Medicine, Creighton University, Omaha, Nebraska; Rheumatology and Bone Diseases Research Group (J.P.B.), CHU de Quebec (CHUL) Research Centre and Department of Medicine, Laval University, Quebec City, Quebec, Canada; United Osteoporosis Centers (C.P.R.), Gainesville, Georgia; New Mexico Clinical Research & Osteoporosis Center (E.M.L.), Albuquerque, New Mexico; Department of Medicine (P.D.M.), Colorado Center for Bone Research, Lakewood, Colorado; Bone & Mineral Research Laboratory (S.D.R.), Henry Ford Hospital, Detroit, Michigan; Department of Medicine (Endocrinology) (D.L.K.), University of British Columbia, Vancouver, British Columbia, Canada; Department of Medicine (R.L.), College of Physicians and Surgeons of Columbia University, New York; Lilly Research Laboratories (J.H.K., J.A.), Eli Lilly and Company, Indianapolis, Indiana; Musculoskeletal and Men's Health (K.A.T., V.A.R.), Lilly USA, LLC, Indianapolis, Indiana; Research and Development - Bio-Medicines (B.J.), Eli Lilly Canada Inc., Toronto, Ontario, Canada
| | - John H Krege
- Regional Bone Center (D.W.D., H.Z., R.L.), Helen Hayes Hospital, West Haverstraw, New York; Department of Pathology and Cell Biology (D.W.D.), College of Physicians and Surgeons of Columbia University, New York; Department of Medicine (R.R.R.), Division of Endocrinology, School of Medicine, Creighton University, Omaha, Nebraska; Rheumatology and Bone Diseases Research Group (J.P.B.), CHU de Quebec (CHUL) Research Centre and Department of Medicine, Laval University, Quebec City, Quebec, Canada; United Osteoporosis Centers (C.P.R.), Gainesville, Georgia; New Mexico Clinical Research & Osteoporosis Center (E.M.L.), Albuquerque, New Mexico; Department of Medicine (P.D.M.), Colorado Center for Bone Research, Lakewood, Colorado; Bone & Mineral Research Laboratory (S.D.R.), Henry Ford Hospital, Detroit, Michigan; Department of Medicine (Endocrinology) (D.L.K.), University of British Columbia, Vancouver, British Columbia, Canada; Department of Medicine (R.L.), College of Physicians and Surgeons of Columbia University, New York; Lilly Research Laboratories (J.H.K., J.A.), Eli Lilly and Company, Indianapolis, Indiana; Musculoskeletal and Men's Health (K.A.T., V.A.R.), Lilly USA, LLC, Indianapolis, Indiana; Research and Development - Bio-Medicines (B.J.), Eli Lilly Canada Inc., Toronto, Ontario, Canada
| | - Jahangir Alam
- Regional Bone Center (D.W.D., H.Z., R.L.), Helen Hayes Hospital, West Haverstraw, New York; Department of Pathology and Cell Biology (D.W.D.), College of Physicians and Surgeons of Columbia University, New York; Department of Medicine (R.R.R.), Division of Endocrinology, School of Medicine, Creighton University, Omaha, Nebraska; Rheumatology and Bone Diseases Research Group (J.P.B.), CHU de Quebec (CHUL) Research Centre and Department of Medicine, Laval University, Quebec City, Quebec, Canada; United Osteoporosis Centers (C.P.R.), Gainesville, Georgia; New Mexico Clinical Research & Osteoporosis Center (E.M.L.), Albuquerque, New Mexico; Department of Medicine (P.D.M.), Colorado Center for Bone Research, Lakewood, Colorado; Bone & Mineral Research Laboratory (S.D.R.), Henry Ford Hospital, Detroit, Michigan; Department of Medicine (Endocrinology) (D.L.K.), University of British Columbia, Vancouver, British Columbia, Canada; Department of Medicine (R.L.), College of Physicians and Surgeons of Columbia University, New York; Lilly Research Laboratories (J.H.K., J.A.), Eli Lilly and Company, Indianapolis, Indiana; Musculoskeletal and Men's Health (K.A.T., V.A.R.), Lilly USA, LLC, Indianapolis, Indiana; Research and Development - Bio-Medicines (B.J.), Eli Lilly Canada Inc., Toronto, Ontario, Canada
| | - Kathleen A Taylor
- Regional Bone Center (D.W.D., H.Z., R.L.), Helen Hayes Hospital, West Haverstraw, New York; Department of Pathology and Cell Biology (D.W.D.), College of Physicians and Surgeons of Columbia University, New York; Department of Medicine (R.R.R.), Division of Endocrinology, School of Medicine, Creighton University, Omaha, Nebraska; Rheumatology and Bone Diseases Research Group (J.P.B.), CHU de Quebec (CHUL) Research Centre and Department of Medicine, Laval University, Quebec City, Quebec, Canada; United Osteoporosis Centers (C.P.R.), Gainesville, Georgia; New Mexico Clinical Research & Osteoporosis Center (E.M.L.), Albuquerque, New Mexico; Department of Medicine (P.D.M.), Colorado Center for Bone Research, Lakewood, Colorado; Bone & Mineral Research Laboratory (S.D.R.), Henry Ford Hospital, Detroit, Michigan; Department of Medicine (Endocrinology) (D.L.K.), University of British Columbia, Vancouver, British Columbia, Canada; Department of Medicine (R.L.), College of Physicians and Surgeons of Columbia University, New York; Lilly Research Laboratories (J.H.K., J.A.), Eli Lilly and Company, Indianapolis, Indiana; Musculoskeletal and Men's Health (K.A.T., V.A.R.), Lilly USA, LLC, Indianapolis, Indiana; Research and Development - Bio-Medicines (B.J.), Eli Lilly Canada Inc., Toronto, Ontario, Canada
| | - Boris Janos
- Regional Bone Center (D.W.D., H.Z., R.L.), Helen Hayes Hospital, West Haverstraw, New York; Department of Pathology and Cell Biology (D.W.D.), College of Physicians and Surgeons of Columbia University, New York; Department of Medicine (R.R.R.), Division of Endocrinology, School of Medicine, Creighton University, Omaha, Nebraska; Rheumatology and Bone Diseases Research Group (J.P.B.), CHU de Quebec (CHUL) Research Centre and Department of Medicine, Laval University, Quebec City, Quebec, Canada; United Osteoporosis Centers (C.P.R.), Gainesville, Georgia; New Mexico Clinical Research & Osteoporosis Center (E.M.L.), Albuquerque, New Mexico; Department of Medicine (P.D.M.), Colorado Center for Bone Research, Lakewood, Colorado; Bone & Mineral Research Laboratory (S.D.R.), Henry Ford Hospital, Detroit, Michigan; Department of Medicine (Endocrinology) (D.L.K.), University of British Columbia, Vancouver, British Columbia, Canada; Department of Medicine (R.L.), College of Physicians and Surgeons of Columbia University, New York; Lilly Research Laboratories (J.H.K., J.A.), Eli Lilly and Company, Indianapolis, Indiana; Musculoskeletal and Men's Health (K.A.T., V.A.R.), Lilly USA, LLC, Indianapolis, Indiana; Research and Development - Bio-Medicines (B.J.), Eli Lilly Canada Inc., Toronto, Ontario, Canada
| | - Valerie A Ruff
- Regional Bone Center (D.W.D., H.Z., R.L.), Helen Hayes Hospital, West Haverstraw, New York; Department of Pathology and Cell Biology (D.W.D.), College of Physicians and Surgeons of Columbia University, New York; Department of Medicine (R.R.R.), Division of Endocrinology, School of Medicine, Creighton University, Omaha, Nebraska; Rheumatology and Bone Diseases Research Group (J.P.B.), CHU de Quebec (CHUL) Research Centre and Department of Medicine, Laval University, Quebec City, Quebec, Canada; United Osteoporosis Centers (C.P.R.), Gainesville, Georgia; New Mexico Clinical Research & Osteoporosis Center (E.M.L.), Albuquerque, New Mexico; Department of Medicine (P.D.M.), Colorado Center for Bone Research, Lakewood, Colorado; Bone & Mineral Research Laboratory (S.D.R.), Henry Ford Hospital, Detroit, Michigan; Department of Medicine (Endocrinology) (D.L.K.), University of British Columbia, Vancouver, British Columbia, Canada; Department of Medicine (R.L.), College of Physicians and Surgeons of Columbia University, New York; Lilly Research Laboratories (J.H.K., J.A.), Eli Lilly and Company, Indianapolis, Indiana; Musculoskeletal and Men's Health (K.A.T., V.A.R.), Lilly USA, LLC, Indianapolis, Indiana; Research and Development - Bio-Medicines (B.J.), Eli Lilly Canada Inc., Toronto, Ontario, Canada
| |
Collapse
|
65
|
Tsuchie H, Miyakoshi N, Kasukawa Y, Nishi T, Abe H, Segawa T, Shimada Y. The effect of teriparatide to alleviate pain and to prevent vertebral collapse after fresh osteoporotic vertebral fracture. J Bone Miner Metab 2016; 34:86-91. [PMID: 25773046 DOI: 10.1007/s00774-014-0646-y] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 12/11/2014] [Indexed: 10/23/2022]
Abstract
Vertebral fracture is often seen in osteoporotic patients. Teriparatide is expected to promote bone union. Therefore, we evaluated the action of vertebral collapse prevention by administering teriparatide to vertebral fracture patients. Thirty-four patients with fresh vertebral fracture (48 vertebrae) participated in this study. They were administered either teriparatide (daily 20 µg/day or weekly 56.5 µg/week) or risedronate (17.5 mg/week): ten patients (20 vertebrae) received teriparatide daily (Daily group), 11 patients (15 vertebrae) received teriparatide weekly (Weekly group), and 13 patients (14 vertebrae) received risedronate (RIS group). We compared some laboratory examination items, visual analogue scale (VAS) of low back pain, vertebral collapse rate and local kyphotic angle, and the cleft frequency. In addition, we evaluated 22 vertebral fracture patients (24 vertebrae) who did not take any osteoporotic medicines (Control group). There was no significant difference in any of the scores at the start of treatment. At 8 and 12 weeks after the initial visit, VAS scores in the Daily and Weekly groups were significantly lower than in the RIS group (p < 0.05). At 8 and 12 weeks, the vertebral collapse rate and local kyphotic angle in the Daily group were significantly lower than in the RIS and Control groups (p < 0.01 and p < 0.05, respectively), and those in the Weekly group were significantly lower than in the Control group (p < 0.05). The cleft frequency in the Daily group was significantly lower than in the RIS group (p < 0.05). Teriparatide is promising for the prevention of vertebral collapse progression after vertebral fracture.
Collapse
Affiliation(s)
- Hiroyuki Tsuchie
- Division of Orthopedic Surgery, Nakadori General Hospital, 3-15, Misono-cho, Minami-dori, Akita, 010-8577, Japan.
| | - Naohisa Miyakoshi
- Department of Orthopedic Surgery, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Yuji Kasukawa
- Department of Orthopedic Surgery, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| | - Tomio Nishi
- Ugo Municipal Hospital, 44-5 Otomichi, Nishimonai, Ugo, 012-1131, Japan
| | - Hidekazu Abe
- Ugo Municipal Hospital, 44-5 Otomichi, Nishimonai, Ugo, 012-1131, Japan
| | - Toyohito Segawa
- Ugo Municipal Hospital, 44-5 Otomichi, Nishimonai, Ugo, 012-1131, Japan
| | - Yoichi Shimada
- Department of Orthopedic Surgery, Akita University Graduate School of Medicine, 1-1-1 Hondo, Akita, 010-8543, Japan
| |
Collapse
|
66
|
Pacifici R. T cells, osteoblasts, and osteocytes: interacting lineages key for the bone anabolic and catabolic activities of parathyroid hormone. Ann N Y Acad Sci 2015; 1364:11-24. [PMID: 26662934 DOI: 10.1111/nyas.12969] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Osteoimmunology is a field of research dedicated to the study of the interactions between the immune system and bone. Among the cells of the immune system that regulate bone turnover and the responsiveness of bone cells to calciothropic hormones are bone marrow T lymphocytes. T cells secrete osteoclastogenic cytokines such as RANKL and TNF-α, as well as factors that stimulate bone formation, one of which is Wnt10b. In addition, T cells regulate the differentiation and life span of stromal cells (SCs) and their responsiveness to parathyroid hormone (PTH) via costimulatory molecules expressed on their surface. The conditioning effect of T cells on SCs is inherited by the osteoblastic and osteocytic progeny of SCs. As a result, osteoblastic cells of T cell-deficient mice have functional characteristics different from corresponding cells of T cell-replete mice. These differences include the ratio of RANKL/OPG produced in response to continuous PTH treatment, and the osteoblastogenic response to intermittent PTH treatment. This article reviews the evidence indicating that the effects of PTH are mediated not only by osteoblasts and osteocytes but also by T cells.
Collapse
Affiliation(s)
- Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, and Immunology and Molecular Pathogenesis Program, Emory University, Atlanta, Georgia
| |
Collapse
|
67
|
Ominsky MS, Brown DL, Van G, Cordover D, Pacheco E, Frazier E, Cherepow L, Higgins-Garn M, Aguirre JI, Wronski TJ, Stolina M, Zhou L, Pyrah I, Boyce RW. Differential temporal effects of sclerostin antibody and parathyroid hormone on cancellous and cortical bone and quantitative differences in effects on the osteoblast lineage in young intact rats. Bone 2015; 81:380-391. [PMID: 26261096 DOI: 10.1016/j.bone.2015.08.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 06/16/2015] [Accepted: 08/06/2015] [Indexed: 12/13/2022]
Abstract
Sclerostin antibody (Scl-Ab) and parathyroid hormone (PTH) are bone-forming agents that have different modes of action on bone, although a study directly comparing their effects has not been conducted. The present study investigated the comparative quantitative effects of these two bone-forming agents over time on bone at the organ, tissue, and cellular level; specifically, at the level of the osteoblast (Ob) lineage in adolescent male and female rats. Briefly, eight-week old male and female Sprague-Dawley rats were administered either vehicle, Scl-Ab (3 or 50mg/kg/week subcutaneously), or human PTH (1-34) (75 μg/kg/day subcutaneously) for 4 or 26 weeks. The 50mg/kg Scl-Ab and the PTH dose were those used in the respective rat lifetime pharmacology studies. Using robust stereological methods, we compared the effects of these agents specifically at the level of the Ob lineage in vertebrae from female rats. Using RUNX2 or nestin immunostaining, location, and morphology, the total number of osteoprogenitor subpopulations, Ob, and lining cells were estimated using the fractionator or proportionator estimators. Density estimates were also calculated referent to total bone surface, total Ob surface, or total marrow volume. Scl-Ab generally effected greater increases in cancellous and cortical bone mass than PTH, correlating with higher bone formation rates (BFR) at 4 weeks in the spine and mid-femur without corresponding increases in bone resorption indices. The increases in vertebral BFR/BS at 4 weeks attenuated with continued treatment to a greater extent with Scl-Ab than with PTH. At 4 weeks, both Scl-Ab and PTH effected equivalent increases in total Ob number (Ob.N). Ob density on the formative surfaces (Ob.N/Ob.S) remained similar across groups while mineral apposition rate (MAR) was significantly higher with Scl-Ab at week 4, reflecting an increase in individual Ob vigor relative to vehicle and PTH. After 26 weeks, Scl-Ab maintained BFR/BS with fewer Ob and lower Ob.N/Ob.S by increasing the Ob footprint (bone surface area occupied by an Ob) and increasing MAR, compared with PTH. The lower Ob.N and Ob.N/Ob.S with Scl-Ab at 26 weeks were associated with decreased osteoprogenitor numbers compared with both vehicle and PTH, an effect not evident at week 4. Osteoprogenitor numbers were generally positively correlated with Ob.N across groups and timepoints, suggesting dynamic coordination between the progenitor and Ob populations. The time-dependent reductions in subpopulations of the Ob lineage with Scl-Ab may be integral to the greater attenuation or self-regulation of bone formation observed at the vertebra, as PTH required more Ob at the formative site with correlative increased numbers of progenitors compared with Scl-Ab indicating potentially greater stimulus for progenitor pool proliferation or differentiation.
Collapse
Affiliation(s)
- Michael S Ominsky
- Department of Metabolic Disorders, Amgen Inc., Thousand Oaks, CA, USA
| | | | - Gwyneth Van
- Department of Comparative Biology and Safety Sciences, Amgen Inc., Thousand Oaks, CA, USA
| | - David Cordover
- Department of Comparative Biology and Safety Sciences, Amgen Inc., Thousand Oaks, CA, USA
| | - Efrain Pacheco
- Department of Comparative Biology and Safety Sciences, Amgen Inc., Thousand Oaks, CA, USA
| | - Emily Frazier
- Department of Comparative Biology and Safety Sciences, Amgen Inc., Thousand Oaks, CA, USA
| | - Linda Cherepow
- Department of Comparative Biology and Safety Sciences, Amgen Inc., Thousand Oaks, CA, USA
| | - Marnie Higgins-Garn
- Department of Comparative Biology and Safety Sciences, Amgen Inc., Thousand Oaks, CA, USA
| | - J Ignacio Aguirre
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Thomas J Wronski
- Department of Physiological Sciences, University of Florida, Gainesville, FL, USA
| | - Marina Stolina
- Department of Metabolic Disorders, Amgen Inc., Thousand Oaks, CA, USA
| | - Lei Zhou
- Global Biostatistical Science, Amgen Inc., Thousand Oaks, CA, USA
| | - Ian Pyrah
- Department of Comparative Biology and Safety Sciences, Amgen Inc., Thousand Oaks, CA, USA
| | - Rogely Waite Boyce
- Department of Comparative Biology and Safety Sciences, Amgen Inc., Thousand Oaks, CA, USA.
| |
Collapse
|
68
|
Kim KM, Rhee Y, Kwon YD, Kwon TG, Lee JK, Kim DY. Medication Related Osteonecrosis of the Jaw: 2015 Position Statement of the Korean Society for Bone and Mineral Research and the Korean Association of Oral and Maxillofacial Surgeons. J Bone Metab 2015; 22:151-65. [PMID: 26713306 PMCID: PMC4691589 DOI: 10.11005/jbm.2015.22.4.151] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 11/29/2015] [Accepted: 11/30/2015] [Indexed: 12/28/2022] Open
Abstract
Bisphosphonates are the most widely prescribed drugs for the treatment of osteoporosis, and are also used in malignant bone metastases, multiple myeloma, and Paget's disease, and provide therapeutic efficacy on those diseases. However, it was reported that occurrence of osteonecrosis of the jaw (ONJ) could be related with bisphosphonate exposures, and there have been many cases regarding this issue. Therefore, a clearer definition and treatment guidelines were needed for this disease. The American Society for Bone and Mineral Research (ASBMR) and American Association of Oral and Maxillofacial Surgeons (AAOMS) reported statements on bisphosphonate-related ONJ (BRONJ), and a revised version was recently presented. In the revised edition, the diagnosis BRONJ was changed to medication-related ONJ (MRONJ), which reflects a consideration of the fact that ONJ also occurs for denosumab, a bone resorption inhibitor of the receptor activator of nuclear factor-kappa B ligand (RANKL) antibody family, and bevacizumab, an anti-angiogenesis inhibitor. In 2009, a statement on ONJ was also reported locally by a relevant organization, which has served as basis for clinical treatment in Korea. In addition to the new official stance of the AAOMS and ASBMR, with an increasing pool of ONJ clinical experience, a revised version of the 2009 local statement is needed. As such, the Korean Society for Bone and Mineral Research (KSBMR) and the Korean Association of Oral and Maxillofacial Surgeons (KAOMS) have collectively formed a committee for the preparation of an official statement on MRONJ, and have reviewed recent local and international data to propose guidelines customized for the local Korean situation.
Collapse
Affiliation(s)
- Kyoung Min Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Yumie Rhee
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Yong-Dae Kwon
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Kyung Hee University, Seoul, Korea
| | - Tae-Geon Kwon
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Kyungpook National University, Daegu, Korea
| | - Jeong Keun Lee
- Division of Oral and Maxillofacial Surgery, Department of Dentistry, Ajou University School of Medicine, Suwon, Korea
| | - Deog-Yoon Kim
- Department of Nuclear Medicine, School of Medicine, Kyung Hee University, Seoul, Korea
| |
Collapse
|
69
|
Compston J. Emerging therapeutic concepts for muscle and bone preservation/building. Bone 2015; 80:150-156. [PMID: 26453503 DOI: 10.1016/j.bone.2015.04.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 03/24/2015] [Accepted: 04/07/2015] [Indexed: 01/16/2023]
Abstract
Loss of muscle or bone mass occurs with ageing, immobility and in association with a variety of systemic diseases. The interaction of these two processes is most evident in the major contribution of falls to the risk of fractures in the elderly population. Exercise and nutrition are key common physiological variables that allow for preservation or formation of greater muscle or bone mass. However, although several pharmacological approaches have the potential to benefit both muscle and bone health, for example vitamin D, selective androgen receptor modulators and ghrelin mimetics, clinical trials with appropriate primary outcomes are lacking. Conventional approaches to address muscle loss are being extended to include stem cell biology and conserved molecular mechanisms of atrophy/hypertrophy. Pharmacological interventions to reduce fracture risk are exploring new mechanisms of action, in particular the uncoupling of bone resorption and formation. Emerging key issues for clinical trial design include adequate phenotyping of patients (personalised medicine), optimisation of the physiological background (multimodal approach) and the use of meaningful and robust outcomes relevant to daily clinical practice. At present, effective treatments that combine beneficial effects on both muscle and bone are lacking, although this is an important target for the future. This review therefore considers current and developing strategies to improve muscle function and bone strength in separate sections.
Collapse
Affiliation(s)
- Juliet Compston
- University of Cambridge School of Clinical Medicine, Department of Medicine, Addenbrooke's Hospital, Hills Road, Cambridge UK CB2 0QQ.
| |
Collapse
|
70
|
|
71
|
Dong Y, Li Y, Huang C, Gao K, Weng X. Systemic application of teriparatide for steroid induced osteonecrosis in a rat model. BMC Musculoskelet Disord 2015; 16:163. [PMID: 26163144 PMCID: PMC4499203 DOI: 10.1186/s12891-015-0589-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 05/18/2015] [Indexed: 12/15/2022] Open
Abstract
Background Steroid associated osteonecrosis is difficult to treat. Teriparatide, as the only one bone anabolic drug, has achieved very promising effect in osteoporosis and other bone skeletal diseases. We carried out this animal study to evaluate the effect of subcutaneous injection of teriparatide for the steroid induced femoral head necrosis in a rat model. Methods 24 Sprague–Dawley male adult rats were included in the study. All the rats were randomized into 4 groups: 18 rats from LPS/MPS group, LPS/MPS + PTH group and LPS/MPS + NS group were given lipopolysaccharide (20 μg/kg) and methylprednisolone (40 mg/kg) to establish the steroid induced osteonecrosis model. 6 rats from NS group only received normal saline. 4 weeks later, All the rats in LPS/MPS group and NS group were sacrificed and the femoral heads were harvested. After that, the 6 rats in the LPS/MPS + PTH group received subcutaneous injection of 20 μg/kg teriparatide and LPS/MPS + NS group only received equal amount of normal saline. After 4 weeks, the serum bone marker was tested and the femoral head were harvested. Micro-CT and histological examination were performed to compare the incidence of osteonecrosis and trabeculae parameters for the femoral head. Results At 4 weeks, rats in LPS/MPS group showed significant osteonecrosis by histological examination (83.3 %) which suggested successful steroid induced osteronecrosis animal models were established. After the treatment of 4 weeks, the LPS/MPS + PTH group showed significant lower incidence rate of osteonecrosis compared with the LPS/MPS + NS group (16.7 % vs.75 %, P < 0.05). The micro CT examination showed higher bone volume/total volume, trabecula thickness and bone mineral density in the LPS/MPS + PTH group compared with the LPS/MPS + NS group. The serum osteocalcin was a little higher in the LPS/MPS + PTH group (4.54 ± 1.61vs.3.58 ± 1.81, P = 0.358), but it didn’t reach a statistical significance. Conclusions Systemic application of teriparatide for steroid induced osteonecrosis in rats showed a beneficial effect. This may be one promising therapy for early stage osteonecrosis.
Collapse
Affiliation(s)
- Yulei Dong
- Deparment of Orthopaedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 100730, Beijing, China.
| | - Yulong Li
- Deparment of Orthopaedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 100730, Beijing, China.
| | - Cheng Huang
- Deparment of Orthopaedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 100730, Beijing, China.
| | - Kai Gao
- Institute of laboratory animal sciences, Chinese Academy of Medical Science and Peking Union Medical College, 100021, Beijing, China.
| | - Xisheng Weng
- Deparment of Orthopaedic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 100730, Beijing, China.
| |
Collapse
|
72
|
Whitmarsh T, Treece GM, Gee AH, Poole KES. Mapping Bone Changes at the Proximal Femoral Cortex of Postmenopausal Women in Response to Alendronate and Teriparatide Alone, Combined or Sequentially. J Bone Miner Res 2015; 30:1309-18. [PMID: 25639838 DOI: 10.1002/jbmr.2454] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Revised: 12/16/2014] [Accepted: 01/10/2015] [Indexed: 11/06/2022]
Abstract
Combining antiresorptive and anabolic drugs for osteoporosis may be a useful strategy to prevent hip fractures. Previous studies comparing the effects of alendronate (ALN) and teriparatide (TPTD) alone, combined or sequentially using quantitative computed tomography (QCT) in postmenopausal women have not distinguished cortical bone mineral density (CBMD) from cortical thickness (CTh) effects, nor assessed the distribution and extent of more localized changes. In this study a validated bone mapping technique was used to examine the cortical and endocortical trabecular changes in the proximal femur resulting from an 18-month course of ALN or TPTD. Using QCT data from a different clinical trial, the global and localized changes seen following a switch to TPTD after an 18-month ALN treatment or adding TPTD to the ALN treatment were compared. Ct.Th increased (4.8%, p < 0.01) and CBMD decreased (-4.5%, p < 0.01) in the TPTD group compared to no significant change in the ALN group. A large Ct.Th increase could be seen for the switch group (2.8%, p < 0.01) compared to a significantly smaller increase for the add group (1.5%, p < 0.01). CBMD decreased significantly for the switch group (-3.9%, p < 0.01) and was significantly different from no significant change in the add group. Ct.Th increases were shown to be significantly greater for the switch group compared to the add group at the load bearing regions. This study provides new insights into the effects of ALN and TPTD combination therapies on the cortex of the proximal femur and supports the hypothesis of an increased bone remodeling by TPTD being mitigated by ALN.
Collapse
Affiliation(s)
| | - Graham M Treece
- Department of Engineering, University of Cambridge, Cambridge, UK
| | - Andrew H Gee
- Department of Engineering, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
73
|
Yang X, Ricciardi BF, Dvorzhinskiy A, Brial C, Lane Z, Bhimani S, Burket JC, Hu B, Sarkisian AM, Ross FP, van der Meulen MCH, Bostrom MPG. Intermittent Parathyroid Hormone Enhances Cancellous Osseointegration of a Novel Murine Tibial Implant. J Bone Joint Surg Am 2015; 97:1074-83. [PMID: 26135074 PMCID: PMC4574908 DOI: 10.2106/jbjs.n.01052] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Long-term fixation of uncemented joint implants requires early mechanical stability and implant osseointegration. To date, osseointegration has been unreliable and remains a major challenge in cementless total knee arthroplasty. We developed a murine model in which an intra-articular proximal tibial titanium implant with a roughened stem can be loaded through the knee joint. Using this model, we tested the hypothesis that intermittent injection of parathyroid hormone (iPTH) would increase proximal tibial cancellous osseointegration. METHODS Ten-week-old female C57BL/6 mice received a subcutaneous injection of PTH (40 μg/kg/day) or a vehicle (n = 45 per treatment group) five days per week for six weeks, at which time the baseline group was killed (n = 6 per treatment group) and an implant was inserted into the proximal part of the tibiae of the remaining mice. Injections were continued until the animals were killed at one week (n = 7 per treatment group), two weeks (n = 14 per treatment group), or four weeks (n = 17 per treatment group) after implantation. Outcomes included peri-implant bone morphology as analyzed with micro-computed tomography (microCT), osseointegration percentage and bone area fraction as shown with backscattered electron microscopy, cellular composition as demonstrated by immunohistochemical analysis, and pullout strength as measured with mechanical testing. RESULTS Preimplantation iPTH increased the epiphyseal bone volume fraction by 31.6%. When the data at post-implantation weeks 1, 2, and 4 were averaged for the iPTH-treated mice, the bone volume fraction was 74.5% higher in the peri-implant region and 168% higher distal to the implant compared with the bone volume fractions in the same regions in the vehicle-treated mice. Additionally, the trabecular number was 84.8% greater in the peri-implant region and 74.3% greater distal to the implant. Metaphyseal osseointegration and bone area fraction were 28.1% and 70.1% higher, respectively, in the iPTH-treated mice than in the vehicle-treated mice, and the maximum implant pullout strength was 30.9% greater. iPTH also increased osteoblast and osteoclast density by 65.2% and 47.0%, respectively, relative to the values in the vehicle group, when the data at post-implantation weeks 1 and 2 were averaged. CONCLUSIONS iPTH increased osseointegration, cancellous mass, and the strength of the bone-implant interface. CLINICAL RELEVANCE Our murine model is an excellent platform on which to study biological enhancement of cancellous osseointegration.
Collapse
Affiliation(s)
- Xu Yang
- Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021. E-mail address for X. Yang:
| | - Benjamin F Ricciardi
- Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021. E-mail address for X. Yang:
| | - Aleksey Dvorzhinskiy
- Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021. E-mail address for X. Yang:
| | - Caroline Brial
- Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021. E-mail address for X. Yang:
| | - Zachary Lane
- Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021. E-mail address for X. Yang:
| | - Samrath Bhimani
- Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021. E-mail address for X. Yang:
| | - Jayme C Burket
- Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021. E-mail address for X. Yang:
| | - Bin Hu
- Department of Biomaterials and Biomimetics, New York University College of Dentistry, 345 East 24th Street, New York, NY 10010
| | - Alexander M Sarkisian
- Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021. E-mail address for X. Yang:
| | - F Patrick Ross
- Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021. E-mail address for X. Yang:
| | | | - Mathias P G Bostrom
- Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021. E-mail address for X. Yang:
| |
Collapse
|
74
|
Birkhold AI, Razi H, Weinkamer R, Duda GN, Checa S, Willie BM. Monitoring in vivo (re)modeling: a computational approach using 4D microCT data to quantify bone surface movements. Bone 2015; 75:210-21. [PMID: 25746796 DOI: 10.1016/j.bone.2015.02.027] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 02/06/2015] [Accepted: 02/25/2015] [Indexed: 10/23/2022]
Abstract
Bone undergoes continual damage repair and structural adaptation to changing external loads with the aim of maintaining skeletal integrity throughout life. The ability to monitor bone (re)modeling would allow for a better understanding in how various pathologies and interventions affect bone turnover and subsequent bone strength. To date, however, current methods to monitor bone (re)modeling over time and in space are limited. We propose a novel method to visualize and quantify bone turnover, based on in vivo microCT imaging and a 4D computational approach. By in vivo tracking of spatially correlated formation and resorption sites over time it classifies bone restructuring into (re)modeling sequences, the spatially and temporally linked sequences of formation, resorption and quiescent periods on the bone surface. The microCT based method was validated using experimental data from an in vivo mouse tibial loading model and ex vivo data of the mouse tibia. In this application, the method allows the visualization of time-resolved cortical (re)modeling and the quantification of short-term and long-term modeling on the endocortical and periosteal surface at the mid-diaphysis of loaded and control mice tibiae. Both short-term and long-term modeling processes, independent formation and resorption events, could be monitored and modeling (spatially not correlated formation and resorption) and remodeling (resorption followed by new formation at the same site) could be distinguished on the bone surface. This novel method that combines in vivo microCT with a computational approach is a powerful tool to monitor bone turnover in animal models now and is waiting to be applied to human patients in the near future.
Collapse
Affiliation(s)
- Annette I Birkhold
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany.
| | - Hajar Razi
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany.
| | - Richard Weinkamer
- Max Planck Institute of Colloids and Interfaces, Department of Biomaterials, Science Park Potsdam-Golm, 14424 Potsdam, Germany.
| | - Georg N Duda
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany; Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany.
| | - Sara Checa
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Bettina M Willie
- Julius Wolff Institute, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|
75
|
Seeman E, Martin TJ. Co-administration of antiresorptive and anabolic agents: a missed opportunity. J Bone Miner Res 2015; 30:753-64. [PMID: 25736531 DOI: 10.1002/jbmr.2496] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 02/13/2015] [Accepted: 02/28/2015] [Indexed: 01/02/2023]
Abstract
Co-administration of antiresorptive and anabolic therapies has appeal because these treatments target the two main abnormalities in bone remodeling responsible for bone loss and microstructural deterioration. Antiresorptives reduce the number of basic multicellular units (BMUs) remodeling bone and reduce the volume of bone each BMU resorbs. Intermittent parathyroid hormone (PTH) increases the volume of bone formed by existing BMUs and those generated by PTH administration. PTH also increases bone formation by stimulating the differentiation, maturation, and longevity of osteoblast lineage cells residing upon quiescent bone surfaces. Despite these rationally targeted actions, enthusiasm for this approach waned when combined therapy blunted the increase in areal bone mineral density (aBMD) relative to that produced by PTH. Although many studies have since reported additive effects of combined therapy, whatever the aBMD result (blunting, additive, or null), these outcomes give little, if any, insight into changes in bone's material composition or microstructure and give misleading information concerning the net effects on bone strength. Combined therapy remains a potentially valuable approach to therapy. Because studies of antifracture efficacy comparing combined with single therapy are unlikely to be performed in humans, efforts should be directed toward improving methods of quantifying the net effects of combined therapy on bone's material composition, microarchitecture, and strength.
Collapse
Affiliation(s)
- Ego Seeman
- Departments of Medicine and Endocrinology, Austin Health, University of Melbourne, Melbourne, Australia
| | | |
Collapse
|
76
|
|
77
|
Silva BC, Bilezikian JP. Parathyroid hormone: anabolic and catabolic actions on the skeleton. Curr Opin Pharmacol 2015; 22:41-50. [PMID: 25854704 DOI: 10.1016/j.coph.2015.03.005] [Citation(s) in RCA: 351] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 03/24/2015] [Indexed: 12/16/2022]
Abstract
Parathyroid hormone (PTH) is essential for the maintenance of calcium homeostasis through, in part, its actions to regulate bone remodeling. While PTH stimulates both bone formation and bone resorption, the duration and periodicity of exposure to PTH governs the net effect on bone mass, that is whether it is catabolic or anabolic. PTH receptor signaling in osteoblasts and osteocytes can increase the RANKL/OPG ratio, increasing both osteoclast recruitment and osteoclast activity, and thereby stimulating bone resorption. In contrast, PTH-induced bone formation is explained, at least in part, by its ability to downregulate SOST/sclerostin expression in osteocytes, permitting the anabolic Wnt signaling pathway to proceed. The two modes of administration of PTH, that is, continuous vs. intermittent, can regulate, in bone cells, different sets of genes; alternatively, the same sets of genes exposed to PTH in sustained vs. transient way, will favor bone resorption or bone formation, respectively. This article reviews the effects of PTH on bone cells that lead to these dual catabolic and anabolic actions on the skeleton.
Collapse
Affiliation(s)
- Barbara C Silva
- Santa Casa de Belo Horizonte and Felicio Rocho Hospital, Division of Endocrinology, Brazil
| | - John P Bilezikian
- Metabolic Bone Diseases Unit, Division of Endocrinology, Department of Medicine, College of Physicians and Surgeons, Columbia University, United States.
| |
Collapse
|
78
|
The genome as pharmacopeia: Association of genetic dose with phenotypic response. Biochem Pharmacol 2015; 94:229-40. [DOI: 10.1016/j.bcp.2015.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Revised: 02/12/2015] [Accepted: 02/12/2015] [Indexed: 11/21/2022]
|
79
|
Robinson JW, Li JY, Walker LD, Tyagi AM, Reott MA, Yu M, Adams J, Weitzmann MN, Pacifici R. T cell-expressed CD40L potentiates the bone anabolic activity of intermittent PTH treatment. J Bone Miner Res 2015; 30:695-705. [PMID: 25359628 PMCID: PMC4376617 DOI: 10.1002/jbmr.2394] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 08/27/2014] [Accepted: 10/28/2014] [Indexed: 01/01/2023]
Abstract
T cells are known to potentiate the bone anabolic activity of intermittent parathyroid hormone (iPTH) treatment. One of the involved mechanisms is increased T cell secretion of Wnt10b, a potent osteogenic Wnt ligand that activates Wnt signaling in stromal cells (SCs). However, additional mechanisms might play a role, including direct interactions between surface receptors expressed by T cells and SCs. Here we show that iPTH failed to promote SC proliferation and differentiation into osteoblasts (OBs) and activate Wnt signaling in SCs of mice with a global or T cell-specific deletion of the T cell costimulatory molecule CD40 ligand (CD40L). Attesting to the relevance of T cell-expressed CD40L, iPTH induced a blunted increase in bone formation and failed to increase trabecular bone volume in CD40L(-/-) mice and mice with a T cell-specific deletion of CD40L. CD40L null mice exhibited a blunted increase in T cell production of Wnt10b and abrogated CD40 signaling in SCs in response to iPTH treatment. Therefore, expression of the T cell surface receptor CD40L enables iPTH to exert its bone anabolic activity by activating CD40 signaling in SCs and maximally stimulating T cell production of Wnt10b.
Collapse
Affiliation(s)
- Jerid W Robinson
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University, Atlanta, GA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
80
|
Periosteum: characteristic imaging findings with emphasis on radiologic-pathologic comparisons. Skeletal Radiol 2015; 44:321-38. [PMID: 25269751 DOI: 10.1007/s00256-014-1976-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 07/21/2014] [Accepted: 07/28/2014] [Indexed: 02/02/2023]
Abstract
The periosteum covers most bone structures. It has an outer fibrous layer and an inner cambial layer that exhibits osteogenic activity. The periosteum is a dynamic structure that plays a major role in bone modeling and remodeling under normal conditions. In several disorders such as infections, benign and malignant tumors, and systemic diseases, the osteogenic potential of the periosteum is stimulated and new bone is produced. The newly formed bone added onto the surface of the cortex adopts various configurations depending on the modalities and pace of bone production. Our aim here is to describe the anatomy, histology, and physiology of the periosteum and to review the various patterns of periosteal reaction with emphasis on relations between radiological and histopathological findings. A careful evaluation of the periosteal reaction and appearance of the underlying cortex, in combination with the MRI, clinical, and laboratory data, provides valuable information on lesion duration and aggressiveness, thereby assisting in the etiological diagnosis and optimizing patient management. A solid reaction strongly suggests a benign and slow-growing process that gives the bone enough time to wall off the lesion. Single lamellar reactions occur in acute and usually benign diseases. Multilamellar reactions are associated with intermediate aggressiveness and a growth rate close to the limit of the walling-off capabilities of the bone. Spiculated, interrupted, and complex combined reactions carry the worst prognosis, as they occur in the most aggressive and fast-growing diseases: the periosteum attempts to create new bone but is overwhelmed and may be breached.
Collapse
|
81
|
Sugiyama T, Kim YT, Oda H. Osteoporosis therapy: a novel insight from natural homeostatic system in the skeleton. Osteoporos Int 2015; 26:443-7. [PMID: 25288445 DOI: 10.1007/s00198-014-2923-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 09/25/2014] [Indexed: 01/22/2023]
Abstract
The skeleton normally responds to mechanical environment to maintain the resulting elastic deformation (strain) of bone, while increased bone strength by an osteoporosis drug results in decreased bone strain. Thus, it can be hypothesized that the effect of osteoporosis therapy is limited by natural homeostatic system in the skeleton. This logic is consistent with the fact that there exists a powerful effect that returns bone mass to its pre-treatment level after the withdrawal of treatment with osteoporosis agents. The present hypothesis provides a new significant insight into the mechanisms by which osteoporosis drugs improve bone fragility. Here we briefly discuss the effects of teriparatide, romosozumab, and odanacatib on bones in animals and humans.
Collapse
Affiliation(s)
- T Sugiyama
- Department of Orthopaedic Surgery, Saitama Medical University, 38 Morohongo, Moroyama, Saitama, 350-0495, Japan,
| | | | | |
Collapse
|
82
|
Grosso MJ, Courtland HW, Yang X, Sutherland JP, Stoner K, Nguyen J, Fahlgren A, Ross FP, van der Meulen MCH, Bostrom MP. Intermittent PTH administration and mechanical loading are anabolic for periprosthetic cancellous bone. J Orthop Res 2015; 33:163-73. [PMID: 25408434 PMCID: PMC4776647 DOI: 10.1002/jor.22748] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The purpose of this study was to determine the individual and combined effects on periprosthetic cancellous bone of intermittent parathyroid hormone administration (iPTH) and mechanical loading at the cellular, molecular, and tissue levels. Porous titanium implants were inserted bilaterally on the cancellous bone of adult rabbits beneath a loading device attached to the distal lateral femur. The left femur received a sham loading device. The right femur was loaded daily, and half of the rabbits received daily PTH. Periprosthetic bone was evaluated up to 28 days for gene expression, histology, and µCT analysis. Loading and iPTH increased bone mass by a combination of two mechanisms: (1) Altering cell populations in a pro-osteoblastic/anti-adipocytic direction, and (2) controlling bone turnover by modulating the RANKL-OPG ratio. At the tissue level, BV/TV increased with both loading (+53%, p < 0.05) and iPTH (+54%, p < 0.05). Combined treatment showed only small additional effects at the cellular and molecular levels that corresponded to a small additive effect on bone volume (+13% compared to iPTH alone, p > 0.05). This study suggests that iPTH and loading are potential therapies for enhancing periprosthetic bone formation. The elucidation of the cellular and molecular response may help further enhance the combined therapy and related targeted treatment strategies.
Collapse
Affiliation(s)
- Matthew J. Grosso
- Laboratory for Mineralized Tissue Research, Hospital for Special Surgery, 535 East 70 Street, New York, NY 10021,Cleveland Clinic Lerner College of Medicine, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195
| | - Hayden-William Courtland
- Laboratory for Mineralized Tissue Research, Hospital for Special Surgery, 535 East 70 Street, New York, NY 10021
| | - Xu Yang
- Laboratory for Mineralized Tissue Research, Hospital for Special Surgery, 535 East 70 Street, New York, NY 10021
| | - James P. Sutherland
- Laboratory for Mineralized Tissue Research, Hospital for Special Surgery, 535 East 70 Street, New York, NY 10021
| | - Kirsten Stoner
- Laboratory for Mineralized Tissue Research, Hospital for Special Surgery, 535 East 70 Street, New York, NY 10021
| | - Joseph Nguyen
- Laboratory for Mineralized Tissue Research, Hospital for Special Surgery, 535 East 70 Street, New York, NY 10021
| | - Anna Fahlgren
- Laboratory for Mineralized Tissue Research, Hospital for Special Surgery, 535 East 70 Street, New York, NY 10021,Department for Clinical and Experimental Medicine, Linkoping University, SE-581 83 Linkoping, Sweden
| | - F. Patrick Ross
- Laboratory for Mineralized Tissue Research, Hospital for Special Surgery, 535 East 70 Street, New York, NY 10021
| | - Marjolein C. H. van der Meulen
- Laboratory for Mineralized Tissue Research, Hospital for Special Surgery, 535 East 70 Street, New York, NY 10021,Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York 14853
| | - Mathias P. Bostrom
- Laboratory for Mineralized Tissue Research, Hospital for Special Surgery, 535 East 70 Street, New York, NY 10021
| |
Collapse
|
83
|
Comparison between recombinant human parathyroid hormone (1–34) and elcatonin in treatment of primary osteoporosis. ASIAN PAC J TROP MED 2015; 8:79-84. [DOI: 10.1016/s1995-7645(14)60192-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 10/10/2014] [Accepted: 11/15/2014] [Indexed: 11/21/2022] Open
|
84
|
Sugiyama T, Torio T, Sato T, Matsumoto M, Kim YT, Oda H. Improvement of skeletal fragility by teriparatide in adult osteoporosis patients: a novel mechanostat-based hypothesis for bone quality. Front Endocrinol (Lausanne) 2015; 6:6. [PMID: 25688232 PMCID: PMC4311704 DOI: 10.3389/fendo.2015.00006] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 01/13/2015] [Indexed: 12/17/2022] Open
Affiliation(s)
- Toshihiro Sugiyama
- Department of Orthopaedic Surgery, Saitama Medical University, Saitama, Japan
- *Correspondence:
| | - Tetsuya Torio
- Department of Orthopaedic Surgery, Saitama Medical University, Saitama, Japan
| | - Tsuyoshi Sato
- Department of Oral and Maxillofacial Surgery, Saitama Medical University, Saitama, Japan
| | - Masahito Matsumoto
- Division of Functional Genomics and Systems Medicine, Research Center for Genomic Medicine, Saitama Medical University, Saitama, Japan
| | - Yoon Taek Kim
- Department of Orthopaedic Surgery, Saitama Medical University, Saitama, Japan
| | - Hiromi Oda
- Department of Orthopaedic Surgery, Saitama Medical University, Saitama, Japan
| |
Collapse
|
85
|
Amugongo SK, Yao W, Jia J, Dai W, Lay YAE, Jiang L, Harvey D, Zimmermann EA, Schaible E, Dave N, Ritchie RO, Kimmel DB, Lane NE. Effect of sequential treatments with alendronate, parathyroid hormone (1-34) and raloxifene on cortical bone mass and strength in ovariectomized rats. Bone 2014; 67:257-68. [PMID: 25016965 PMCID: PMC4157684 DOI: 10.1016/j.bone.2014.04.033] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 04/03/2014] [Accepted: 04/16/2014] [Indexed: 01/10/2023]
Abstract
UNLABELLED Anti-resorptive and anabolic agents are often prescribed for the treatment of osteoporosis continuously or sequentially for many years. However their impact on cortical bone quality and bone strength is not clear. METHODS Six-month old female rats were either sham operated or ovariectomized (OVX). OVX rats were left untreated for two months and then were treated with vehicle (Veh), hPTH (1-34) (PTH), alendronate (Aln), or raloxifene (Ral) sequentially for three month intervals, for a total of three periods. Mid-tibial cortical bone architecture, mass, mineralization, and strength were measured on necropsy samples obtained after each period. Bone indentation properties were measured on proximal femur necropsy samples. RESULTS Eight or more months of estrogen deficiency in rats resulted in decreased cortical bone area and thickness. Treatment with PTH for 3months caused the deposition of endocortical lamellar bone that increased cortical bone area, thickness, and strength. These improvements were lost when PTH was withdrawn without followup treatment, but were maintained for the maximum times tested, six months with Ral and three months with Aln. Pre-treatment with anti-resorptives was also somewhat successful in ultimately preserving the additional endocortical lamellar bone formed under PTH treatment. These treatments did not affect bone indentation properties. SUMMARY Sequential therapy that involved both PTH and anti-resorptive agents was required to achieve lasting improvements in cortical area, thickness, and strength in OVX rats. Anti-resorptive therapy, either prior to or following PTH, was required to preserve gains attributable to an anabolic agent.
Collapse
Affiliation(s)
- Sarah K Amugongo
- Musculoskeletal Research Unit, Department of Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Wei Yao
- Musculoskeletal Research Unit, Department of Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Junjing Jia
- Musculoskeletal Research Unit, Department of Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Weiwei Dai
- Musculoskeletal Research Unit, Department of Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Yu-An E Lay
- Musculoskeletal Research Unit, Department of Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Li Jiang
- Musculoskeletal Research Unit, Department of Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Danielle Harvey
- Division of Biostatistics, Department of Public Health Sciences, University of California, Davis, CA 95616, USA
| | - Elizabeth A Zimmermann
- Materials Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Eric Schaible
- Experimental Systems Group, Advanced Light Source, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Neil Dave
- Materials Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Robert O Ritchie
- Musculoskeletal Research Unit, Department of Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA; Department of Materials Science and Engineering, University of California, Berkeley, CA 94720, USA
| | - Donald B Kimmel
- Osteoporosis Research Center, School of Medicine, Creighton University, Omaha, NE 68131, USA
| | - Nancy E Lane
- Musculoskeletal Research Unit, Department of Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA.
| |
Collapse
|
86
|
Eriksen EF, Keaveny TM, Gallagher ER, Krege JH. Literature review: The effects of teriparatide therapy at the hip in patients with osteoporosis. Bone 2014; 67:246-56. [PMID: 25053463 DOI: 10.1016/j.bone.2014.07.014] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 06/19/2014] [Accepted: 07/08/2014] [Indexed: 01/24/2023]
Abstract
Teriparatide is a skeletal anabolic treatment for patients with osteoporosis at high risk for fracture. Because adequate clinical trials have not yet been conducted to assess the efficacy of teriparatide for reducing the risk of hip fracture, we review here the literature regarding how treatment with teriparatide affects the hip in patients with osteoporosis. Teriparatide increases cancellous bone volume, improves bone architecture, and - uniquely among osteoporosis treatments - increases cortical thickness and cortical porosity. By bone scan and positron emission tomography, teriparatide increases bone formation throughout the skeleton, including the hip. Consistent with these findings, studies using dual-energy X-ray absorptiometry and quantitative computed tomography for longitudinal assessment of changes at the hip have consistently shown increases in areal and volumetric bone mineral density, cortical thickness, and finite element-estimated hip strength in patients treated with teriparatide. Finally, in clinical fracture-outcome trials, treatment with teriparatide has been shown to reduce the risk of nonvertebral fracture, a composite endpoint that includes hip fracture. Taken together, this body of evidence suggests that teriparatide positively affects the hip in patients with osteoporosis.
Collapse
Affiliation(s)
- Erik F Eriksen
- Department of Endocrinology, Oslo University Hospital, Pb 49596 Nydalen, N-0424 Oslo, Norway.
| | - Tony M Keaveny
- University of California, Berkeley, Departments of Mechanical Engineering and Bioengineering, 6175 Etcheverry Hall, MC 1740, Berkeley, CA 94720, USA.
| | - Eileen R Gallagher
- inVentiv Health Clinical, 504 Carnegie Center, Princeton, NJ 08540, USA.
| | - John H Krege
- Lilly USA, LLC, Lilly Technology Center South, Drop Code 5028 Indianapolis, IN 46221, USA.
| |
Collapse
|
87
|
Niimi R, Kono T, Nishihara A, Hasegawa M, Matsumine A, Kono T, Sudo A. Determinants associated with bone mineral density increase in response to daily teriparatide treatment in patients with osteoporosis. Bone 2014; 66:26-30. [PMID: 24909538 DOI: 10.1016/j.bone.2014.05.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/14/2014] [Accepted: 05/29/2014] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Several factors associated with bone mineral density (BMD) increase are reported with daily teriparatide treatment, but there has been no systematic analysis to summarize these associations. The purpose of this study was to investigate the clinical determinants associated with BMD increase to daily teriparatide treatment. METHODS This was a retrospective study. We performed an analysis of 306 patients diagnosed with osteoporosis. Teriparatide was administered at 20μg/day for 12months. The primary efficacy measure was a change in lumbar spine (LS) BMD from baseline at 12months. To determine the response variables of BMD changes, we investigated the clinical determinants using univariate and multivariate analyses. RESULTS There was a 9.8±8.2% increase in LS BMD after 12months. Prior bisphosphonate treatment and baseline procollagen type I N-terminal propeptide (PINP) concentration were significantly associated with LS BMD absolute response by univariate analyses. In the multiple regression model, patients with higher baseline PINP concentration had a significantly greater LS BMD absolute increase. Prior bisphosphonate use lost its correlation in the multiple regression models. CONCLUSION Our results showed that baseline PINP concentration was a useful predictor of LS BMD absolute increase regardless of prior treatment.
Collapse
Affiliation(s)
- Rui Niimi
- Department of Orthopaedic Surgery, Tomidahama Hospital, 26-14, Tomidahama-cho, Yokkaichi-city, Mie 510-8008, Japan.
| | - Toshibumi Kono
- Department of Orthopaedic Surgery, Tomidahama Hospital, 26-14, Tomidahama-cho, Yokkaichi-city, Mie 510-8008, Japan.
| | - Atsushi Nishihara
- Department of Orthopaedic Surgery, Tomidahama Hospital, 26-14, Tomidahama-cho, Yokkaichi-city, Mie 510-8008, Japan.
| | - Masahiro Hasegawa
- Department of Orthopaedic Surgery, Mie University, Graduate School of Medicine, 2-174, Edobashi, Tsu-city, Mie 514-8507, Japan.
| | - Akihiko Matsumine
- Department of Orthopaedic Surgery, Mie University, Graduate School of Medicine, 2-174, Edobashi, Tsu-city, Mie 514-8507, Japan.
| | - Toshihiko Kono
- Department of Orthopaedic Surgery, Tomidahama Hospital, 26-14, Tomidahama-cho, Yokkaichi-city, Mie 510-8008, Japan.
| | - Akihiro Sudo
- Department of Orthopaedic Surgery, Mie University, Graduate School of Medicine, 2-174, Edobashi, Tsu-city, Mie 514-8507, Japan.
| |
Collapse
|
88
|
Pennypacker BL, Chen CM, Zheng H, Shih MS, Belfast M, Samadfam R, Duong LT. Inhibition of cathepsin K increases modeling-based bone formation, and improves cortical dimension and strength in adult ovariectomized monkeys. J Bone Miner Res 2014; 29:1847-58. [PMID: 24591096 DOI: 10.1002/jbmr.2211] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 02/17/2014] [Accepted: 02/18/2014] [Indexed: 12/21/2022]
Abstract
Treatment with the cathepsin K (CatK) inhibitor odanacatib (ODN) protects against bone loss and maintains normal biomechanical properties in the spine and hip of ovariectomized (OVX) preclinical models. Here, we characterized the effects of ODN on the dynamics of cortical modeling and remodeling, and dimension and strength of the central femur in adult OVX-rhesus monkeys. Animals were treated with vehicle or ODN (6 or 30 mg/kg, once per day [q.d., p.o.]) in prevention mode for 21 months. Calcein and tetracycline double-labeling were given at 12 and 21 months, and the femoral cross-sections were subjected to dynamic histomorphometric and cement line analyses. ODN treatment significantly increased periosteal and endocortical bone formation (BFR/BS), accompanied with an increase in endocortical mineralizing surface (102%, p < 0.01) with the 6 mg/kg dose. ODN at both doses reduced remodeling hemiosteon numbers by 51% and 66% (p < 0.05), respectively, and ODN 30 mg/kg numerically reduced activation frequency without affecting wall thickness. On the same endocortical surface, ODN increased all modeling-based parameters, while reducing intracortical remodeling, consistent with the observed no treatment effects on cortical porosity. ODN 30 mg/kg markedly increased cortical thickness (CtTh, p < 0.001) and reduced marrow area (p < 0.01). Lastly, ODN treatment increased femoral structural strength (p < 0.001). Peak load was positively correlated with the increases in bone mineral content (BMC) (r(2) = 0.9057, p < 0.0001) and CtTh (r2 = 0.6866, p < 0.0001). Taken together, by reducing cortical remodeling-based and stimulating modeling-based bone formation, ODN significantly improved cortical dimension and strength in OVX monkeys. This novel mechanism of CatK inhibition in stimulating cortical formation suggests that ODN represents a novel therapeutic approach for the treatment of osteoporosis.
Collapse
|
89
|
Kleerekoper M, Greenspan SL, Lewiecki EM, Miller PD, Kendler DL, Maricic M, Keaveny TM, Kopperdahl DL, Ruff VA, Wan X, Janos B, Krohn K. Assessing the Effects of Teriparatide Treatment on Bone Mineral Density, Bone Microarchitecture, and Bone Strength. J Bone Joint Surg Am 2014; 96:e90. [PMID: 24897747 PMCID: PMC6948798 DOI: 10.2106/jbjs.l.01757] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND To gain insight into how teriparatide affects various bone health parameters, we assessed the effects of teriparatide treatment with use of standard DXA (dual x-ray absorptiometry) technology and two newer technologies, high-resolution MRI (magnetic resonance imaging) and finite element analysis of quantitative CT (computed tomography) scans. METHODS In this phase-4, open-label study, postmenopausal women with severe osteoporosis received 20 μg/day of teriparatide. Assessments included (1) changes in areal BMD (bone mineral density) (in g/cm2) at the radius, spine, and hip on DXA, (2) changes in volumetric BMD (in mg/cm3) at the spine and hip on quantitative CT scans, (3) changes in bone microarchitecture at the radius on high-resolution MRI, (4) estimated changes in spine and hip strength according to finite element analysis of quantitative CT scans, (5) changes in bone turnover markers in serum, and (6) safety. RESULTS Thirty-five subjects were enrolled; thirty completed eighteen months and twenty-five completed an optional six-month extension. No significant changes were observed for the primary outcome, high-resolution MRI at the distal aspect of the radius. At month eighteen, the least-squares mean percentage change from baseline in total volumetric BMD at the spine was 10.05% (95% confidence interval [CI], 6.83% to 13.26%; p < 0.001), and estimated spine strength increased 17.43% (95% CI, 12.09% to 22.76%; p < 0.001). Total volumetric BMD at the hip increased 2.22% (95% CI, 0.37% to 4.06%; p = 0.021), and estimated hip strength increased 2.54% (95% CI, 0.06% to 5.01%; p = 0.045). Areal BMD increased at the lumbar spine and femoral neck, was unchanged for the total hip and at the distalmost aspect of the radius, and decreased at a point one-third of the distance between the wrist and elbow. Bone turnover markers increased at months three, six, and twenty-four (all p < 0.05). No unexpected adverse events were observed. CONCLUSIONS High-resolution MRI failed to identify changes in bone microarchitecture at the distal aspect of the radius, a non-weight-bearing site that may not be suitable for assessing effects of an osteoanabolic agent. Teriparatide increased areal BMD at the spine and femoral neck and volumetric BMD at the spine and hip. Estimated vertebral and femoral strength also increased. These findings and increases in bone turnover markers through month twenty-four are consistent with the known osteoanabolic effect of teriparatide. LEVEL OF EVIDENCE Therapeutic Level IV. See Instructions for Authors for a complete description of levels of evidence.
Collapse
Affiliation(s)
- Michael Kleerekoper
- Division of Endocrinology, Department of Medicine, University of Toledo, 3000 Arlington Avenue, Toledo, OH 43614. E-mail address:
| | - Susan L Greenspan
- Osteoporosis Treatment and Prevention Center, University of Pittsburgh, 3459 Fifth Avenue, 4th Floor, Pittsburgh, PA 15213
| | - E Michael Lewiecki
- New Mexico Clinical Research & Osteoporosis Center, 300 Oak Street N.E., Albuquerque, NM 87106
| | - Paul D Miller
- Colorado Center for Bone Research, University of Colorado Health Sciences Center, 3190 South Wadsworth Boulevard, Lakewood, CO 80227
| | - David L Kendler
- University of British Columbia, 150-943 West Broadway, Vancouver, BC V5Z 4E1, Canada
| | - Michael Maricic
- Catalina Pointe Rheumatology, 7520 North Oracle Road, Suite 100, Tucson, AZ 85724
| | - Tony M Keaveny
- Departments of Mechanical Engineering and Bioengineering, 6175 Etcheverry Hall, MC 1740, University of California, Berkeley, CA 94720-1740
| | | | - Valerie A Ruff
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285
| | - Xiaohai Wan
- Novartis Pharmaceuticals Corporation, One Health Plaza East, East Hanover, NJ 07936
| | - Boris Janos
- Eli Lilly Canada Inc., 3650 Danforth Avenue, Toronto, ON M1N 2E8, Canada
| | - Kelly Krohn
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285
| |
Collapse
|
90
|
Ominsky MS, Niu QT, Li C, Li X, Ke HZ. Tissue-level mechanisms responsible for the increase in bone formation and bone volume by sclerostin antibody. J Bone Miner Res 2014; 29:1424-30. [PMID: 24967455 DOI: 10.1002/jbmr.2152] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Bone formation can be remodeling-based (RBF) or modeling-based (MBF), the former coupled to bone resorption and the latter occurring directly on quiescent surfaces. Unlike osteoanabolic therapies such as parathyroid hormone (PTH) 1-34 that increase bone remodeling and thus both formation and resorption, sclerostin antibody (Scl-Ab) increases bone formation while decreasing bone resorption. With this unique profile, we tested our hypothesis that Scl-Ab primarily elicited MBF by examining bones from Scl-Ab–treated ovariectomized (OVX) rats and male cynomolgus monkeys (cynos). Histomorphometry was performed to quantify and characterize bone surfaces in OVX rats administered vehicle or Scl-Ab (25 mg/kg) subcutaneously (sc) twice/week for 5 weeks and in adolescent cynos administered vehicle or Scl-Ab (30 mg/kg) sc every 2 weeks for 10 weeks. Fluorochrome-labeled surfaces in L2 vertebra and femur endocortex (cynos only) were considered to be MBF or RBF based on characteristics of their associated cement lines. In OVX rats, Scl-Ab increased MBF by eightfold (from 7% to 63% of bone surface, compared to vehicle). In cynos, Scl-Ab markedly increased MBF on trabecular (from 0.6% to 34%) and endocortical surfaces (from 7% to 77%) relative to vehicle. Scl-Ab did not significantly affect RBF in rats or cynos despite decreased resorption surface in both species. In cynos, Scl-Ab resulted in a greater proportion of RBF and MBF containing sequential labels from week 2, indicating an increase in the lifespan of the formative site. This extended formation period was associated with robust increases in the percent of new bone volume formed. These results demonstrate that Scl-Ab increased bone volume by increasing MBF and prolonged the formation period at both modeling and remodeling sites while reducing bone resorption. Through these unique effects on bone formation and resorption, Scl-Ab may prove to be an effective therapeutic to rapidly increase bone mass in diseases such as osteoporosis.
Collapse
Affiliation(s)
| | - Qing-Tian Niu
- Department of Metabolic Disorders; Amgen Inc.; Thousand Oaks CA USA
| | - Chaoyang Li
- Department of Metabolic Disorders; Amgen Inc.; Thousand Oaks CA USA
| | - Xiaodong Li
- Department of Metabolic Disorders; Amgen Inc.; Thousand Oaks CA USA
| | - Hua Zhu Ke
- Department of Metabolic Disorders; Amgen Inc.; Thousand Oaks CA USA
| |
Collapse
|
91
|
Ng KW, Martin TJ. New therapeutics for osteoporosis. Curr Opin Pharmacol 2014; 16:58-63. [PMID: 24699340 DOI: 10.1016/j.coph.2014.03.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 03/12/2014] [Accepted: 03/12/2014] [Indexed: 12/11/2022]
Abstract
Two new approaches for the treatment of osteoporosis are summarized, each having arisen out of important new discoveries in bone biology. Odanacatib (ODN) inhibits the enzyme, cathepsin K, that is essential for the resorbing activity of osteoclasts. It is effective in preventing ovariectomy-induced bone loss in preclinical studies, and a phase II clinical study has shown inhibition of resorption sustained over five years. Outcome of a phase III study is awaited. The finding from mouse and human genetics that Wnt signaling is a powerful inducer of bone formation led to developments aimed at enhancing this pathway. Of the several approaches towards this, the most advanced is with a neutralizing antibody against sclerostin, the osteocyte-derived inhibitor of Wnt signaling. Preclinical studies show a powerful bone anabolic effect, and a clinical phase II study shows dose-dependent increases in bone formation and decreases in bone resorption markers.
Collapse
Affiliation(s)
- Kong Wah Ng
- University of Melbourne, Department of Medicine, St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy 3065, Victoria, Australia
| | - T John Martin
- University of Melbourne, Department of Medicine, St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy 3065, Victoria, Australia.
| |
Collapse
|
92
|
Martin TJ. Bone biology and anabolic therapies for bone: current status and future prospects. J Bone Metab 2014; 21:8-20. [PMID: 24707463 PMCID: PMC3970295 DOI: 10.11005/jbm.2014.21.1.8] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 02/03/2014] [Accepted: 02/03/2014] [Indexed: 12/20/2022] Open
Abstract
Bone is continuously remodelled at many sites asynchronously throughout the skeleton, with bone formation and resorption balanced at these sites to retain bone structure. Negative balance resulting in bone loss and osteoporosis, with consequent fractures, has mainly been prevented or treated by anti-resorptive drugs that inhibit osteoclast formation and/or activity, with new prospects now of anabolic treatments that restore bone that has been lost. The anabolic effectiveness of parathyroid hormone has been established, and an exciting new prospect is presented of neutralising antibody against the osteocyte protein, sclerostin. The cellular actions of these two anabolic treatments differ, and the mechanisms will need to be kept in mind in devising their best use. On present evidence it seems likely that treatment with either of these anabolic agents will need to be followed by anti-resorptive treatment in order to maintain bone that has been restored. No matter how effective anabolic therapies for the skeleton become, it seems highly likely that there will be a continuing need for safe, effective anti-resorptive drugs.
Collapse
Affiliation(s)
- T John Martin
- Department of Medicine, St Vincent's Institute of Medical Research, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
93
|
Ma YL, Zeng QQ, Chiang AY, Burr D, Li J, Dobnig H, Fahrleitner-Pammer A, Michalská D, Marin F, Pavo I, Stepan JJ. Effects of teriparatide on cortical histomorphometric variables in postmenopausal women with or without prior alendronate treatment. Bone 2014; 59:139-47. [PMID: 24269280 DOI: 10.1016/j.bone.2013.11.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 11/05/2013] [Accepted: 11/12/2013] [Indexed: 11/25/2022]
Abstract
Cortical bone, the dominant component of the human skeleton by volume, plays a key role in protecting bones from fracture. We analyzed the cortical bone effects of teriparatide treatment in postmenopausal women with osteoporosis who had previously received long-term alendronate (ALN) therapy or were treatment naïve (TN). Tetracycline-labeled paired iliac crest biopsies obtained from 29 ALN-pretreated and 16 TN women were evaluated for dynamic histomorphometric parameters of bone formation at the periosteal, endocortical and intracortical bone compartments, before and after 24months of teriparatide treatment. At baseline, the frequency of specimens without any endocortical and periosteal tetracycline labeling, and the percentage of quiescent osteons, was higher in the ALN than the TN group. Endocortical and periosteal mineralizing surface (MS/BS%), periosteal bone formation rate (BFR/BS), mineral apposition rate (MAR) and the number of intracortical forming osteons were significantly lower in the ALN-pretreated patients than in the TN group. Following teriparatide treatment, the frequency of endocortical and periosteal unlabeled biopsies decreased; in the ALN-pretreated group the percentage of quiescent osteons decreased and, in contrast, forming and resorbing osteons were increased. Teriparatide treatment resulted in significant increases of MAR in the endocortical, and MS/BS% in the periosteal compartment in the ALN-pretreated group. Most indices of bone formation remained lower in the ALN-pretreated group compared with the TN group at study end. Endocortical wall width was increased in both ALN-pretreated and TN groups. Cortical porosity and cortical thickness were significantly increased in the ALN-pretreated group after teriparatide treatment. Our results suggest that 24months of teriparatide treatment increases cortical bone formation and cortical turnover in patients who were either TN or had previous ALN therapy.
Collapse
Affiliation(s)
- Yanfei L Ma
- Lilly Research Laboratories, Indianapolis, IN, USA.
| | - Qing Q Zeng
- Lilly Research Laboratories, Indianapolis, IN, USA.
| | | | - David Burr
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Jiliang Li
- Department of Biology, Indiana University Purdue University, Indianapolis, IN, USA.
| | - Harald Dobnig
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Medical University of Graz, Graz, Austria.
| | - Astrid Fahrleitner-Pammer
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Medical University of Graz, Graz, Austria.
| | - Dana Michalská
- 3rd Dept. of Internal Medicine, Charles University Faculty of Medicine 1, Prague, Czech Republic.
| | | | - Imre Pavo
- Lilly Research Centre, Windlesham, United Kingdom.
| | - Jan J Stepan
- Institute of Rheumatology, and Charles University Faculty of Medicine 1, Prague, Czech Republic.
| |
Collapse
|
94
|
Li JY, Walker LD, Tyagi AM, Adams J, Weitzmann MN, Pacifici R. The sclerostin-independent bone anabolic activity of intermittent PTH treatment is mediated by T-cell-produced Wnt10b. J Bone Miner Res 2014; 29:43-54. [PMID: 24357520 PMCID: PMC4326235 DOI: 10.1002/jbmr.2044] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Revised: 06/11/2013] [Accepted: 06/29/2013] [Indexed: 12/29/2022]
Abstract
Both blunted osteocytic production of the Wnt inhibitor sclerostin (Scl) and increased T-cell production of the Wnt ligand Wnt10b contribute to the bone anabolic activity of intermittent parathyroid hormone (iPTH) treatment. However, the relative contribution of these mechanisms is unknown. In this study, we modeled the repressive effects of iPTH on Scl production in mice by treatment with a neutralizing anti-Scl antibody (Scl-Ab) to determine the contribution of T-cell-produced Wnt10b to the Scl-independent modalities of action of iPTH. We report that combined treatment with Scl-Ab and iPTH was more potent than either iPTH or Scl-Ab alone in increasing stromal cell production of OPG, osteoblastogenesis, osteoblast life span, bone turnover, bone mineral density, and trabecular bone volume and structure in mice with T cells capable of producing Wnt10b. In T-cell-null mice and mice lacking T-cell production of Wnt10b, combined treatment increased bone turnover significantly more than iPTH or Scl-Ab alone. However, in these mice, combined treatment with Scl-Ab and iPTH was equally effective as Scl-Ab alone in increasing the osteoblastic pool, bone volume, density, and structure. These findings demonstrate that the Scl-independent activity of iPTH on osteoblasts and bone mass is mediated by T-cell-produced Wnt10b. The data provide a proof of concept of a more potent therapeutic effect of combined treatment with iPTH and Scl-Ab than either alone.
Collapse
Affiliation(s)
- Jau-Yi Li
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Lindsey D Walker
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Abdul Malik Tyagi
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University, Atlanta, GA, USA
| | - Jonathan Adams
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University, Atlanta, GA, USA
| | - M Neale Weitzmann
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University, Atlanta, GA, USA
- Atlanta VA Medical Center, Decatur, GA, USA
| | - Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University, Atlanta, GA, USA
- Immunology and Molecular Pathogenesis Program, Emory University, Atlanta, GA, USA
| |
Collapse
|
95
|
Pacifici R. Role of T cells in the modulation of PTH action: physiological and clinical significance. Endocrine 2013; 44:576-82. [PMID: 23729167 PMCID: PMC3815684 DOI: 10.1007/s12020-013-9960-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 04/10/2013] [Indexed: 12/24/2022]
Abstract
Osteoimmunology is new field of research dedicated to the study of the interactions between the immune system and bone. Among the cells of the immune system that regulate bone and hemopoietic cells are T lymphocytes. These cells secrete osteoclastogenic cytokines such as RANKL and TNF, as well as factors that stimulate bone formation and hemopoietic cells, one of which is Wnt10b. This article will review the evidence that T cells are implicated in the mechanism of action of parathyroid hormone (PTH) in bone and on the hemopoietic system.
Collapse
Affiliation(s)
- Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory University School of Medicine, 101 Woodruff Circle, Room 1309, Atlanta, GA, 30322, USA,
| |
Collapse
|
96
|
Kang SY, Deshpande SS, Donneys A, Rodriguez JJ, Nelson NS, Felice PA, Chepeha DB, Buchman SR. Parathyroid hormone reverses radiation induced hypovascularity in a murine model of distraction osteogenesis. Bone 2013; 56:9-15. [PMID: 23643680 PMCID: PMC3758112 DOI: 10.1016/j.bone.2013.04.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 04/19/2013] [Accepted: 04/20/2013] [Indexed: 01/30/2023]
Abstract
BACKGROUND Radiation treatment results in a severe diminution of osseous vascularity. Intermittent parathyroid hormone (PTH) has been shown to have an anabolic effect on osteogenesis, though its impact on angiogenesis remains unknown. In this murine model of distraction osteogenesis, we hypothesize that radiation treatment will result in a diminution of vascularity in the distracted regenerate and that delivery of intermittent systemic PTH will promote angiogenesis and reverse radiation induced hypovascularity. MATERIALS AND METHODS Nineteen Lewis rats were divided into three groups. All groups underwent distraction of the left mandible. Two groups received radiation treatment to the left mandible prior to distraction, and one of these groups was treated with intermittent subcutaneous PTH (60 μg/kg, once daily) beginning on the first day of distraction for a total duration of 21 days. One group underwent mandibular distraction alone, without radiation. After consolidation, the rats were perfused and imaged with micro-CT angiography and quantitative vascular analysis was performed. RESULTS Radiation treatment resulted in a severe diminution of osseous vascularity in the distracted regenerate. In irradiated mandibles undergoing distraction osteogenesis, treatment with intermittent PTH resulted in significant increases in vessel volume fraction, vessel thickness, vessel number, degree of anisotropy, and a significant decrease in vessel separation (p < 0.05). No significant difference in quantitative vascularity existed between the group that was irradiated, distracted and treated with PTH and the group that underwent distraction osteogenesis without radiation treatment. CONCLUSIONS We quantitatively demonstrate that radiation treatment results in a significant depletion of osseous vascularity, and that intermittent administration of PTH reverses radiation induced hypovascularity in the murine mandible undergoing distraction osteogenesis. While the precise mechanism of PTH-induced angiogenesis remains to be elucidated, this report adds a key component to the pleotropic effect of intermittent PTH on bone formation and further supports the potential use of PTH to enhance osseous regeneration in the irradiated mandible.
Collapse
Affiliation(s)
- Stephen Y. Kang
- Craniofacial Research Laboratory, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Health System, 1500 E. Medical Center Dr., 1904 TC / SPC 5312, Ann Arbor, MI, USA
| | - Sagar S. Deshpande
- Craniofacial Research Laboratory, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Alexis Donneys
- Craniofacial Research Laboratory, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Joey J. Rodriguez
- Craniofacial Research Laboratory, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Noah S. Nelson
- Craniofacial Research Laboratory, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Peter A. Felice
- Craniofacial Research Laboratory, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Douglas B. Chepeha
- Craniofacial Research Laboratory, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Otolaryngology-Head and Neck Surgery, University of Michigan Health System, 1500 E. Medical Center Dr., 1904 TC / SPC 5312, Ann Arbor, MI, USA
| | - Steven R. Buchman
- Craniofacial Research Laboratory, University of Michigan Medical School, Ann Arbor, MI, USA
- Section of Plastic Surgery, University of Michigan Health System, 1500 E. Medical Center Dr., F7894 / 5219, Ann Arbor, MI, USA
| |
Collapse
|
97
|
Tsai JN, Uihlein AV, Lee H, Kumbhani R, Siwila-Sackman E, McKay EA, Burnett-Bowie SAM, Neer RM, Leder BZ. Teriparatide and denosumab, alone or combined, in women with postmenopausal osteoporosis: the DATA study randomised trial. Lancet 2013; 382:50-6. [PMID: 23683600 PMCID: PMC4083737 DOI: 10.1016/s0140-6736(13)60856-9] [Citation(s) in RCA: 295] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Osteoporosis medications increase bone-mineral density (BMD) and lower but do not eliminate fracture risk. The combining of anabolic agents with bisphosphonates has not improved efficacy. We compared combined teriparatide and denosumab with both agents alone. METHODS From September, 2009, to January, 2011, we enrolled postmenopausal women with osteoporosis into this randomised, controlled trial. Patients were assigned in a 1:1:1 ratio to receive 20 μg teriparatide daily, 60 mg denosumab every 6 months, or both. BMD was measured at 0, 3, 6, and 12 months. Women who completed at least one study visit after baseline were assessed in a modified intention-to-treat analysis. This trial is registered with ClinicalTrials.gov, number NCT00926380. FINDINGS 94 (94%) of 100 eligible women completed at least one study visit after baseline. At 12 months, posterior-anterior lumbar spine BMD increased more in the combination group (9·1%, [SD 3·9]) than in the teriparatide (6·2% [4·6], p=0·0139) or denosumab (5·5% [3·3], p=0·0005) groups. Femoral-neck BMD also increased more in the combination group (4·2% [3·0]) than in the teriparatide (0·8% [4·1], p=0·0007) and denosumab (2·1% [3·8], p=0·0238) groups, as did total-hip BMD (combination, 4·9% [2·9]; teriparatide, 0·7% [2·7], p<0·0001; denosumab 2·5% [2·6], p=0·0011). INTERPRETATION Combined teriparatide and denosumab increased BMD more than either agent alone and more than has been reported with approved therapies. Combination treatment might, therefore, be useful to treat patients at high risk of fracture. FUNDING Amgen, Eli Lilly, National Center for Research Resources.
Collapse
Affiliation(s)
- Joy N Tsai
- Department of Medicine, Endocrine Unit, Massachusetts General Hospital, Boston, MA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Peng S, Zhang G, Zhang BT, Guo B, He Y, Bakker AJ, Pan X, Zhen W, Hung L, Qin L, Leung WN. The beneficial effect of icaritin on osteoporotic bone is dependent on the treatment initiation timing in adult ovariectomized rats. Bone 2013; 55:230-40. [PMID: 23486180 DOI: 10.1016/j.bone.2013.02.012] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 02/05/2013] [Accepted: 02/06/2013] [Indexed: 02/01/2023]
Abstract
BACKGROUND Epimedium-derived flavonoids (EFs) have a potential to treat established osteoporosis in postmenopausal women. However, one of the main disadvantages of the compound is the high volume and dosage during long-term administration period. Meanwhile, the beneficial effect of EFs on osteoporotic bone depends greatly on the intervention timing. Whether icaritin (ICT), an active molecular compound from EFs, can exert beneficial effect on osteoporotic bone and whether the beneficial effect is also dependent on the intervention timing remain unknown. OBJECTIVE The objective of this study was to evaluate the effect of the early and late ICT treatment on bone turnover markers, trabecular architecture, bone remodeling, biomechanics, colony formation of bone marrow stromal cells and osteoblast, adipocyte and osteoclast-related gene expression in adult ovariectomized rats. METHODS Eighty 9-month-old female rats (n=8/group) were sham-operated (Sham) or ovariectomized (OVX). The OVX rats were subjected to ICT treatment initiation at 1 month (early treatment) and 3 months (late treatment) post-operation, respectively. The vehicle-treated Sham and OVX rats starting at month 1 and month 3 post-operation served as the corresponding controls (Sham and OVX controls) for early and late ICT treatment, respectively. Those Sham and OVX rats sacrificed immediately before early and late ICT treatment served as the pretreatment baseline controls. Both ICT and vehicle treatments lasted for 2 months. The bone turnover markers, trabecular architecture, bone remodeling and bone biomechanical properties were analyzed with biochemistry, microCT, histomorphometry and mechanical testing, respectively. The population of bone marrow stromal cells (BMSCs) and osteoblasts were evaluated with colony formation assays, respectively. The expression levels of osteoblast, adipocyte and osteoclast-related genes in bone marrow were assessed by real-time polymerase chain reaction (PCR), respectively. RESULTS At the tissue level, early ICT treatment remarkably restored the trabecular bone mass, trabecular architecture and bone biomechanical properties towards pretreatment Sham levels, and significantly increased bone formation from pretreatment OVX level and markedly inhibited bone resorption towards pretreatment Sham level, whereas late ICT treatment failed to have any effect. At the cellular and molecular level, early ICT treatment significantly increased the number of osteoblastic colonies and the level of osteoblast-related gene expression compared to pretreatment OVX levels and remarkably decreased adipocyte and osteoclast-related gene expression towards pretreatment Sham levels. Late ICT treatment failed to have beneficial effect on any of these parameters. CONCLUSION ICT can exert anabolic and anti-resorptive effect on osteoporotic bone. The beneficial effect of ICT treatment is dependent on the intervention timing in established osteoporosis induced by estrogen depletion.
Collapse
Affiliation(s)
- Songlin Peng
- Department of Spine Surgery, Shenzhen People's Hospital, Jinan University Second College of Medicine, Shenzhen, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
99
|
Saito H, Takeda S, Amizuka N. Eldecalcitol and calcitriol stimulates 'bone minimodeling,' focal bone formation without prior bone resorption, in rat trabecular bone. J Steroid Biochem Mol Biol 2013; 136:178-82. [PMID: 23069645 DOI: 10.1016/j.jsbmb.2012.10.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 10/02/2012] [Accepted: 10/04/2012] [Indexed: 10/27/2022]
Abstract
Vitamin D is known as a potent stimulator of bone resorption. The active form of vitamin D3, calcitriol (1α,25-dihydroxyvitamin D3), stimulates release of calcium (Ca) from bone in ex vivo organ culture, and treatment with large amounts of an active vitamin D3 analog induces hypercalcemia and bone resorption in mice in vivo. Calcitriol strongly induces both receptor activator of NF-κB ligand (RANKL) and macrophage colony-stimulating factor (M-CSF) in osteoblasts in vitro. On the other hand, it has been reported that active vitamin D3 inhibits bone resorption in various experimental animal models. We previously showed that eldecalcitol [1α,25-dihydroxy-2β-(3-hydroxy-propyloxy)vitamin D3; ED-71] suppresses bone resorption and increases bone mineral density (BMD) to a greater extent than alfacalcidol (1α-hydroxyvitamin D3) in ovariectomized (OVX) rats in vivo. To elucidate the histological events that follow administration of eldecalcitol compared to calcitriol, OVX rats were given either vehicle, eldecalcitol (10, 30, or 90ng/kg), or calcitriol (33.3, 100, 300, or 900ng/kg), and sham-operated control animals were given vehicle, 5-times per week for 12 weeks. The lumbar spine and femur were removed and processed for bone mineral density (BMD) assessments and the femur for histomorphometrical analyzes. Both eldecalcitol and calcitriol increased the lumbar and femoral BMD in a dose dependent manner. Bone histomorphometry revealed that osteoclast surface (Oc.S/BS) and eroded surface (ES/BS) were dose-dependently suppressed in the trabecular region of the femur. Both calcitriol and eldecalcitol dose-dependently stimulated focal bone formation that started without prior bone resorption, a process known as bone minimodeling. Both reduction of bone resorption and stimulation of focal bone formation were more clearly observed in the eldecalcitol-treated rats than in the calcitriol-treated rats. Taken together, these findings suggest that eldecalcitol is a more potent vitamin D3 analog that stimulates focal bone formation (minimodeling) and suppresses bone resorption more strongly than does calcitriol. This article is part of a Special Issue entitled 'Vitamin D Workshop'.
Collapse
Affiliation(s)
- Hitoshi Saito
- Medical Science Department, Chugai Pharmaceutical Co., Ltd., Tokyo, Japan.
| | | | | |
Collapse
|
100
|
Hansen S, Hauge EM, Beck Jensen JE, Brixen K. Differing effects of PTH 1-34, PTH 1-84, and zoledronic acid on bone microarchitecture and estimated strength in postmenopausal women with osteoporosis: an 18-month open-labeled observational study using HR-pQCT. J Bone Miner Res 2013; 28:736-45. [PMID: 23044908 DOI: 10.1002/jbmr.1784] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Revised: 09/17/2012] [Accepted: 10/01/2012] [Indexed: 11/10/2022]
Abstract
Whereas the beneficial effects of intermittent treatment with parathyroid hormone (PTH) (intact PTH 1-84 or fragment PTH 1-34, teriparatide) on vertebral strength is well documented, treatment may not be equally effective in the peripheral skeleton. We used high-resolution peripheral quantitative computed tomography (HR-pQCT) to detail effects on compartmental geometry, density, and microarchitecture as well as finite element (FE) estimated integral strength at the distal radius and tibia in postmenopausal osteoporotic women treated with PTH 1-34 (20 µg sc daily, n = 18) or PTH 1-84 (100 µg sc daily, n = 20) for 18 months in an open-label, nonrandomized study. A group of postmenopausal osteoporotic women receiving zoledronic acid (5 mg infusion once yearly, n = 33) was also included. Anabolic therapy increased cortical porosity in radius (PTH 1-34 32 ± 37%, PTH 1-84 39 ± 32%, both p < 0.001) and tibia (PTH 1-34 13 ± 27%, PTH 1-84 15 ± 22%, both p < 0.001) with corresponding declines in cortical density. With PTH 1-34, increases in cortical thickness in radius (2.0 ± 3.8%, p < 0.05) and tibia (3.8 ± 10.4%, p < 0.01) were found. Trabecular number increased in tibia with both PTH 1-34 (4.2 ± 7.1%, p < 0.05) and PTH 1-84 (5.3 ± 8.3%, p < 0.01). Zoledronic acid did not impact cortical porosity at either site but increased cortical thickness (3.0 ± 3.5%, p < 0.01), total (2.7 ± 2.5%, p < 0.001) and cortical density (1.5 ± 2.0%, p < 0.01) in tibia as well as trabecular volume fraction in radius (2.5 ± 5.1%, p < 0.05) and tibia (2.2 ± 2.2%, p < 0.01). FE estimated bone strength was preserved, but not increased, with PTH 1-34 and zoledronic acid at both sites, whereas it decreased with PTH 1-84 in radius (-2.8 ± 5.8%, p < 0.05) and tibia (-3.9 ± 4.8%, p < 0.001). Conclusively, divergent treatment-specific effects in cortical and trabecular bone were observed with anabolic and zoledronic acid therapy. The finding of decreased estimated strength with PTH 1-84 treatment was surprising and warrants confirmation.
Collapse
Affiliation(s)
- Stinus Hansen
- Department of Endocrinology, Odense University Hospital, Odense, Denmark.
| | | | | | | |
Collapse
|