Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Katja M. Fagerlund is active.

Publication


Featured researches published by Katja M. Fagerlund.


Journal of the National Cancer Institute | 2013

Survival Benefit With Radium-223 Dichloride in a Mouse Model of Breast Cancer Bone Metastasis

Mari I. Suominen; Jukka P. Rissanen; Rami Käkönen; Katja M. Fagerlund; Esa Alhoniemi; Dominik Mumberg; Karl Ziegelbauer; Jussi M. Halleen; Sanna-Maria Käkönen; Arne Scholz

BACKGROUND Bone metastases are associated with increased morbidity and poor prognosis in breast cancer patients. Radium-223 dichloride is a calcium mimetic that localizes to bone, providing targeted therapy for skeletal metastasis. METHODS We investigated the mode of action of radium-223 dichloride using breast cancer cell, osteoclast, and osteoblast cultures as well as a mouse model of breast cancer bone metastasis. A single dose of radium-223 dichloride was used in three different settings mimicking the prevention or treatment of bone metastasis. Disease progression was monitored using fluorescence and radiographic imaging and histological analyses. The effect of radium-223 dichloride alone and in combination with doxorubicin or zoledronic acid on survival of mice was analyzed by Kaplan-Meier methods. All statistical tests used were two-sided. RESULTS Radium-223 dichloride incorporated into bone matrix and inhibited proliferation of breast cancer cells and differentiation of osteoblasts and osteoclasts (all P values < .001) in vitro. In an established bone metastasis setting, radium-223 dichloride prevented tumor-induced cachexia (0/14 vs 7/14 control mice) and decreased osteolysis by 56% and tumor growth by 43% (all P values < .05). Radium-223 dichloride induced double-strand DNA breaks in cancer cells in vivo. Finally, radium-223 dichloride extended survival as a monotherapy (29.2 days, 95% confidence interval [CI] = 26.6 to 31.8 days, P = .039) and in combination with zoledronic acid (31.4 days, 95% CI = 28.8 to 34.0 days, P = .004) or doxorubicin (31.5 days, 95% CI = 29.5 to 33.5 days, P < .001) compared to the vehicle group (24.9 days, 95% CI = 23.4 to 26.4 days). Similar but even more pronounced effects were observed when radium-223 dichloride was administered in a preventive or micrometastatic setting. CONCLUSIONS Our findings strongly support the development of radium-223 dichloride for the treatment of breast cancer patients with or at high risk of developing bone metastases.


Clinical Cancer Research | 2017

Radium-223 Inhibits Osseous Prostate Cancer Growth by Dual Targeting of Cancer Cells and Bone Microenvironment in Mouse Models

Mari I. Suominen; Katja M. Fagerlund; Jukka P. Rissanen; Yvonne Konkol; Jukka Morko; Zhiqi Peng; Esa Alhoniemi; Salla K. Laine; Eva Corey; Dominik Mumberg; Karl Ziegelbauer; Sanna-Maria Käkönen; Jussi M. Halleen; Robert L. Vessella; Arne Scholz

Purpose: Radium-223 dichloride (radium-223, Xofigo), a targeted alpha therapy, is currently used for the treatment of patients with castration-resistant prostate cancer (CRPC) with bone metastases. This study examines the mode-of-action and antitumor efficacy of radium-223 in two prostate cancer xenograft models. Experimental Design: Mice bearing intratibial LNCaP or LuCaP 58 tumors were randomized into groups (n = 12–17) based on lesion grade and/or serum PSA level and administered radium-223 (300 kBq/kg) or vehicle, twice at 4-week intervals. X-rays and serum samples were obtained biweekly. Soft tissue tumors were observed macroscopically at sacrifice. Tibiae were analyzed by gamma counter, micro-CT, autoradiography and histology. Results: Radium-223 inhibited tumor-induced osteoblastic bone growth and protected normal bone architecture, leading to reduced bone volume in LNCaP and abiraterone-resistant LuCaP 58 models. Furthermore, radium-223 resulted in lower PSA values and reduced total tissue and tumor areas, indicating that treatment constrains prostate cancer growth in bone. In addition, radium-223 suppressed abnormal bone metabolic activity as evidenced by decreased number of osteoblasts and osteoclasts and reduced level of the bone formation marker PINP. Mode-of-action studies revealed that radium-223 was deposited in the intratumoral bone matrix. DNA double-strand breaks were induced in cancer cells within 24 hours after radium-223 treatment, and PSA levels were significantly lower 72 hours after treatment, providing further evidence of the antitumor effects. Conclusions: Taken together, radium-223 therapy exhibits a dual targeting mode-of-action that induces tumor cell death and suppresses tumor-induced pathologic bone formation in tumor microenvironment of osseous CRPC growth in mice. Clin Cancer Res; 23(15); 4335–46. ©2017 AACR.


Journal of Bone and Mineral Metabolism | 2009

Improved methods for testing antiresorptive compounds in human osteoclast cultures

Jukka P. Rissanen; Hannele Ylipahkala; Katja M. Fagerlund; Clive Long; H. Kalervo Väänänen; Jussi M. Halleen

We cultured human bone marrow-derived stem cells on bovine bone slices in 96-well plates in the presence of M-CSF and RANKL, allowing them to differentiate into osteoclasts. Secreted TRACP 5b was a useful endpoint measurement to demonstrate effects of inhibitors of osteoclast differentiation in the culture system, reflecting accurately the number of formed osteoclasts. Inhibitors of osteoclast activity were added into the cultures after the differentiation period, and the cultures were continued to allow the formed osteoclasts to resorb bone. CTX values obtained after the resorption period were normalized with TRACP 5b values obtained after the differentiation period, before adding the inhibitors. This normalization prevents false results that could be obtained from the presence of different amounts of osteoclasts in different wells before adding the inhibitors. These results demonstrate that the use of TRACP 5b and CTX allows rapid and reliable testing of antiresorptive compounds in human osteoclast cultures.


bonekey Reports | 2013

Novel perspectives on the transcytotic route in osteoclasts

Mirkka J. Hirvonen; Katja M. Fagerlund; Päivi T. Lakkakorpi; H. Kalervo Väänänen; Mika Mulari

We analyzed the characteristics of degraded bone matrix-delivering vesicles along the transcytotic route from the ruffled border to the functional secretory domain (FSD) in bone-penetrating osteoclasts. Cells of rat or human origin were cultured on bovine bone slices and analyzed via confocal microscopy. Helix pomatia lectin binding indicated that transcytotic vesicles expose aberrant N-acetylgalactosamine glycoconjugates, which is associated with a poor prognosis for a range of metastasizing human adenocarcinomas. Transcytotic vesicles fuse with the autophagosomal compartments and represent raft concentrates. Furthermore, the results of a vertical vesicle analysis suggest that multiple vesicle populations arise from the ruffled border and that some of these vesicles undergo a maturation process along the transcytotic route. Finally, our data suggest that the targeting of these membrane pathways may be determined by a novel F-actin-containing and FSD-circumscribing molecular barrier.


Cancer Research | 2014

Abstract 1803: A new spectrum-selective cathepsin inhibitor, VBY-825, inhibits bone destruction in a syngeneic 5TGM1 multiple myeloma mouse model

Mari I. Suominen; Johanna Tuomela; Esa Alhoniemi; Katja M. Fagerlund; Jukka P. Rissanen; Jussi M. Halleen; Leslie J. Holsinger

Multiple myeloma (MM) is the second most common blood cancer after non Hodgkin lymphoma. It is a monoclonal B-cell neoplasia with clinical hallmarks of multiple osteolytic lesions causing bone pain, fractures and hypercalcemia. Chemo- or radiotherapy may induce remissions, but MM is generally thought to be incurable. Our aim was to observe the effects of a cathepsin inhibitor VBY-825 on bone lesions and tumor burden in the syngeneic 5TGM1 mouse MM model using immunocompetent C57BL/KaLwRij mice. VBY-825 is a potent inhibitor of cathepsins K, L, B, V, and S. 5TGM1 cells were inoculated via tail vein in 7 weeks old female C57BL/KaLwRij mice, which were divided to 4 groups: Control group received vehicle of VBY-825 (5% dextrose 10 ml/kg daily), Control group received bortezomib vehicle (3 ml/kg twice a week), Reference group received bortezomib (0.5 mg/kg twice a week) and Study group received VBY-825 (100 mg/kg daily). Administration of all compounds began one day before tumor cell inoculation and continued until day 34. Disease progression was followed by measuring the serum levels of paraprotein (IgG2b) and TRACP 5b, radiography, and body weight. The animals were sacrificed 5 weeks after inoculation, examined macroscopically, and their bones were collected for histomorphometric analysis. The reference compound bortezomib had no effects on body weight but it delayed the disease progression based on IgG2b measurements. It also decreased the number and total area of osteolytic lesions, but not mean osteolytic lesion area (MOLA). VBY-825 had no effect on body weight or IgG2b level, frequency of soft tissue tumors or intraosseous tumor area. VBY-825 decreased total and MOLA, consistent with inhibited resorption. There was also a trend of increased relative trabecular bone area. Serum TRACP 5b activity in the VBY-825 treated group did not differ from the respective vehicle group, whereas the number of osteoclasts at tumor-bone interface was increased in VBY-825 treated animals. These findings suggest that VBY-825 decreased osteoclast function and resorption activity without decreasing the number of osteoclasts In conclusion, VBY-825 had no effects on tumor growth but it inhibited bone destruction in this mouse model of MM, which is consistent with its potency on cathepsin S and K, which are known to be important in osteoclast-mediated bone resorption. VBY-825 is a promising candidate for the treatment of tumor-associated bone disease. Citation Format: Mari I. Suominen, Johanna Tuomela, Esa Alhoniemi, Katja M. Fagerlund, Jukka P. Rissanen, Jussi M. Halleen, Leslie J. Holsinger. A new spectrum-selective cathepsin inhibitor, VBY-825, inhibits bone destruction in a syngeneic 5TGM1 multiple myeloma mouse model. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1803. doi:10.1158/1538-7445.AM2014-1803


Molecular Cancer Therapeutics | 2015

Abstract A109: Radium-223 dichloride inhibits tumor growth and tumor-induced bone growth in osteoblastic prostate cancer models

Mari I. Suominen; Katja M. Fagerlund; Dominik Mumberg; Karl Ziegelbauer; Sanna-Maria Käkönen; Jussi M. Halleen; Robert L. Vessella

Radium-223 dichloride (radium-223), an alpha particle-emitting calcium-mimetic, improves overall survival in prostate cancer patients with symptomatic bone metastases. We have defined radium-223 mode-of-action and efficacy in two clinically relevant prostate cancer xenograft models demonstrating PSA expression and osteoblastic growth upon intratibial inoculation of cancer cells. Immunocompromised male mice were inoculated with human LNCaP or patient-derived LuCaP 58 prostate cancer cells in the intratibial compartment and subsequently stratified into treatment groups based on lesion grade and/or serum PSA levels. Radium-223 (300 kBq/kg) or vehicle was administered intravenously, two times at 4-week intervals during the experiment. X-rays and serum samples were obtained biweekly and at sacrifice. Soft tissue tumors were examined macroscopically at sacrifice and tissue samples were collected and processed for γ-counter measurements, micro-CT, autoradiography and histology. Radium-223 treatment inhibited tumor-induced osteoblastic bone growth as indicated by reduced bone volume and surface in LNCaP and LuCaP 58 prostate cancer mouse models. In addition, radium-223 treatment suppressed metabolic activity in bone as evidenced by decreased number of osteoblasts and osteoclasts relative to bone surface and reduced levels of the bone formation marker PINP. Radium-223 resulted in lower PSA values as early as two weeks after the first dose, indicating constrained tumor growth following treatment. This phenomenon was further supported by reduced total bone lesion tissue and tumor area in LNCaP and LuCaP 58 models and increased percentage of necrotic tumor area in the LuCaP 58 model in radium-223-treated mice as compared to vehicle-treated mice. Moreover, DNA double-strand breaks were increased in cancer cells 24 hours post radium-223 treatment in the LuCaP 58 model providing further evidence of anti-tumor effects. Radium-223-treated mice exhibited less visceral metastases in the LuCaP 58 model (not significant). Based on autoradiography, radium-223 was deposited in the intratumoral bone matrix and in conjunction with osteoblasts in osteoblastic metastases. Our results demonstrate that radium-223 dichloride is successfully incorporated into the intratumoral bone matrix and inhibits tumor growth in both cell line- and patient-derived osteoblastic prostate cancer metastasis models. Given the α-particle range of 50-80 μm, potent radiation effects on the tumor microenvironment are evident whereas relevant effects on the more distant bone marrow are not expected. Taken together, radium-223 therapy exhibits a dual mode-of-action that impacts tumor growth and tumor-induced bone reaction, both important players in the destructive vicious cycle of osteoblastic bone metastasis in prostate cancer. Citation Format: Mari I. Suominen, Katja M. Fagerlund, Dominik Mumberg, Karl Ziegelbauer, Sanna-Maria Kakonen, Jussi M. Halleen, Robert L. Vessella, Arne Scholz. Radium-223 dichloride inhibits tumor growth and tumor-induced bone growth in osteoblastic prostate cancer models. [abstract]. In: Proceedings of the AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics; 2015 Nov 5-9; Boston, MA. Philadelphia (PA): AACR; Mol Cancer Ther 2015;14(12 Suppl 2):Abstract nr A109.


Cancer Research | 2017

Abstract 5202: Additive benefits of radium-223 dichloride and bortezomib combination in a syngeneic 5TGM1 multiple myeloma mouse model

Mari I. Suominen; Jukka P. Rissanen; Anniina Luostarinen; Katja M. Fagerlund; Birgitta Sjöholm; Esa Alhoniemi; Sanna-Maria Käkönen; Dominik Mumberg; Jussi M. Halleen; Karl Ziegelbauer; Arne Scholz

Radium-223 dichloride (radium-223, Xofigo®), a targeted alpha-therapy, improves overall survival in prostate cancer patients with bone metastases. It inhibits disease progression by reducing tumor growth and tumor-induced pathological bone reaction in breast and prostate cancer mouse models. Radium-223 is actively incorporated into the bone matrix by osteoblasts. Multiple myeloma (MM) is characterized by increased osteoclast and reduced or no osteoblast activity. Bortezomib (Velcade®), a treatment for MM, restores the impaired osteoblast activity in MM. Here, we report the effects of radium-223, bortezomib and their combination on myeloma cell proliferation in vitro and on myeloma bone disease model in mice. Proliferation assays were performed with human plasma cell leukemia (JJN-3, L-363), human MM (LP-1, MOLP-8, RPMI-8226 and OPM-2), and mouse MM (5TGM1) cells. Corresponding in vivo effects were studied in a syngeneic 5TGM1 mouse MM model. Female C57BL/KaLwRij mice (7 weeks old, n=15/group) were inoculated with 5TGM1 cells via tail vein and 26 days later, radium-223 (300 kBq/kg, single iv injection) and/or bortezomib (1 mg/kg ip, twice a week; total of 3 doses) or vehicle control were administered. The development of osteolytic lesions was detected by radiography. Hind limbs were used for histological analyses and total activity measurement was performed by a gamma-counter. TRAP-stained osteoclasts were counted at tumor-bone interface. Bortezomib inhibited proliferation of all cancer cell lines tested at 25 nM (JJN3 and OPM-2 at 2.5 nM) and radium-223 at 0.8 kBq/ml (L-363 and MOLP-8 at 0.2 kBq/ml) concentrations. Additive effects were observed with combination treatment in vitro. The 5TGM1 in vivo model demonstrated that both bortezomib and radium-223 decreased osteolytic lesion area as monotherapy (p In conclusion, radium-223 dichloride (Xofigo®) therapy in combination with bortezomib decreased osteolytic lesion area and almost completely eradicated tumor-associated osteoclasts in a mouse model of myeloma bone disease. Incorporation of radium-223 to bone matrix was improved, possibly via induction of osteoblast activity by bortezomib. These data suggest that combination of radium-223 and bortezomib could be a new effective therapy in MM, which is currently being investigated in a Phase Ib/II trial in patients with early relapsed MM (NCT02928029). Citation Format: Mari I. Suominen, Jukka P. Rissanen, Anniina Luostarinen, Katja M. Fagerlund, Birgitta Sjoholm, Esa Alhoniemi, Sanna-Maria Kakonen, Dominik Mumberg, Jussi M. Halleen, Karl Ziegelbauer, Arne Scholz. Additive benefits of radium-223 dichloride and bortezomib combination in a syngeneic 5TGM1 multiple myeloma mouse model [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 5202. doi:10.1158/1538-7445.AM2017-5202


Cancer Research | 2017

Abstract 4207: Selective drug sensitivity score (DSS) for indolent and aggressive prostate cancer cell lines

Jenni Mäki-Jouppila; Jenni Bernoulli; Johanna Tuomela; Mari I. Suominen; Jussi M. Halleen; Sanna Timonen; Elina Huovari; Katja Suomi; Swapnil Potdar; Päivi Östling; Jani Saarela; Katja M. Fagerlund

Prostate cancer (PC) is the most common malignancy in men and the second leading cause of cancer-related deaths. The majority of the PCs are classified as adenocarcinomas characterized by the expression of androgen receptor (AR) and prostate-specific antigen (PSA). Two of the most commonly used cell lines are LNCaP and PC-3 cells, derived from lymph node and bone metastases, respectively. Also VCaP cells, derived from vertebral metastases, are widely used in prostate cancer research. It has been well established that LNCaP and VCaP cells represent the conventional indolent form of PC expressing AR and PSA and are androgen-dependent. PC-3 cells, on the other hand, do not express AR and PSA, are androgen-independent, and represent the highly aggressive form. The drug sensitivity of the cell lines was assessed by applying a large panel of drugs covering cancer chemotherapeutics and clinically available and emerging drugs including conventional chemotherapy, kinase inhibitors, metabolic modifiers, rapalogs, differentiating/epigenetic modifiers, kinesin inhibitors, apoptotic modulators, NSAIDs, hormone therapy, immunomodulators and HSP inhibitors. A panel of 460 compounds was tested in five concentrations covering a 10.000-fold drug-relevant concentration range in 384-well format. Cells were seeded to pre-drugged plates, followed by cell viability measurements (CellTiter-Glo) after 72 hours. Maximal and minimal responses to drugs were analyzed, the EC50 values were calculated and Drug Sensitivity Score (DSS) was calculated for each drug as a measure of reduced viability. A selective Drug Sensitivity Score (sDSS) was calculated to identify the selective drug response pattern of each three cancer cell lines. As expected, the results indicate that LNCaP and VCaP cells in general were more sensitive to drugs of different categories than PC-3 cells. According to DSS analysis, all three cell lines showed sensitivity to conventional chemotherapy and kinase inhibitors. However, PC-3 cells were more sensitive to kinase inhibitors than conventional chemotherapy. Determining sDSS revealed specific sensitivities of each cell line. LNCaP cells were sensitive to kinase inhibitors, such as mTOR and AKT inhibitors. Also VCaP cells showed selective sensitivity to kinase inhibitors, especially Aurora kinase and IGF1R inhibitors. In addition to kinase inhibitors, VCaP cells were selectively sensitive to HDAC inhibitors. Furthermore, PC-3 cells were sensitive to e.g. CDK inhibitors. We conclude that the cell-based compound screening combined with DSS and sDSS analysis provides a possibility to profile cellular responses to an extensive collection of anti-cancer compounds enabling repurposing of existing drugs to new indications, identification of vulnerabilities in different types of cancer cells and functional investigation of cellular pathways behind drug sensitivity or resistance. Citation Format: Jenni Maki-Jouppila, Jenni Bernoulli, Johanna Tuomela, Mari I. Suominen, Jussi M. Halleen, Sanna Timonen, Elina Huovari, Katja Suomi, Swapnil Potdar, Paivi Ostling, Jani Saarela, Katja M. Fagerlund. Selective drug sensitivity score (DSS) for indolent and aggressive prostate cancer cell lines [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4207. doi:10.1158/1538-7445.AM2017-4207


Cancer Research | 2017

Abstract 3838: Drug sensitivity profile of 5TGM1 murine multiple myeloma cell line emphasizes the translational potential of the syngeneicin vivomodel

Jenni Mäki-Jouppila; Jenni Bernoulli; Mari I. Suominen; Tiina E. Kähkönen; Jussi M. Halleen; Sanna Timonen; Elina Huovari; Katja Suomi; Swapnil Potdar; Maria Nurmi; Päivi Östling; Jani Saarela; Katja M. Fagerlund

Multiple myeloma (MM) is the second most common hematologic malignancy that originates from B-cells (plasma cells) and causes 2% of cancer-related deaths. Symptoms of MM include bone pain caused by multiple osteolytic lesions, pathologic fractures, and hypercalcemia. Typically, MM has a low growth fraction and it is highly dependent on the microenvironment. These properties have made it hard to target by conventional chemotherapy, but could now be exploited by novel stroma-targeting drugs and immunotherapy. These new approaches underline the need for well characterized models with functional immune system and appropriate tumor microenvironment. To gain additional information supporting the use of the syngeneic 5TGM1 murine multiple myeloma model in drug development, we tested drug sensitivity of 5TGM1 cells by screening an extensive panel of drugs. The compound library consisting of 460 compounds included conventional chemotherapy, kinase inhibitors, metabolic modifiers, rapalogs, differentiating/epigenetic modifiers, kinesin inhibitors, apoptotic modulators, NSAIDs, hormone therapy, immunomodulators and HSP inhibitors. The compounds were tested in five concentrations covering a 10.000-fold drug-relevant concentration range in 384-well format. Cells were seeded to plates with a compound library, followed by cell viability measurements (CellTiter-Glo) after 72 hours. Maximal and minimal responses to drugs were analyzed, and the EC50 values were calculated. Drug Sensitivity Score (DSS) was calculated for each drug as a measure of reduced viability. According to DSS analysis, 5TGM1 cells showed sensitivity to conventional chemotherapy, such as antimitotic drugs, and kinase inhibitors, such as MEK1/2 inhibitors. In addition, the cells showed particular sensitivity to several HSP90 inhibitors currently in phase I/II clinical development for MM. Lenalidomide and pomalidomide, efficient in treating multiple myeloma in humans, both gave low DSS value indicating that 5TGM1 cells are not sensitive to these drugs, which is expected because they do not bind to murine form of the target cereblon. In contrast, 5TGM1 cells were highly sensitive to the proteasome inhibitor bortezomib (DSS 32.2), which is currently in clinical use. In conclusion, the murine 5TGM1 cells show sensitivity to various MM drugs used in the clinic and under development. Evaluating the effects of the microenvironment on the growth and drug sensitivity of 5TGM1 cells in vitro and in vivo will be essential. Furthermore, the cell-based compound screening combined with DSS analysis provides a possibility to profile cellular responses to an extensive collection of anti-cancer compounds enabling identification of vulnerabilities in cancer cells and functional investigation of cellular pathways behind drug sensitivity or resistance. Citation Format: Jenni Maki-Jouppila, Jenni Bernoulli, Mari I. Suominen, Tiina Kahkonen, Jussi M. Halleen, Sanna Timonen, Elina Huovari, Katja Suomi, Swapnil Potdar, Maria Nurmi, Paivi Ostling, Jani Saarela, Katja M. Fagerlund. Drug sensitivity profile of 5TGM1 murine multiple myeloma cell line emphasizes the translational potential of the syngeneic in vivo model [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3838. doi:10.1158/1538-7445.AM2017-3838


Cancer Research | 2016

Abstract 364: Validation of human osteoclast cultures for studying the mode-of-action and identification of new compounds with the potential of inhibiting the vicious cycle in osteolytic bone metastases

Jenni Bernoulli; Jussi M. Halleen; Mari I. Suominen; Johanna Tuomela; Jukka P. Rissanen; Katja M. Fagerlund

When tumor cells home to bone microenvironment they secrete factors that stimulate osteoclast formation, which results in increased bone resorption. This in turn increases the release of factors from bone matrix that stimulate the growth of tumor cells, leading to a vicious cycle characterized by extensive bone loss and enhanced tumor growth in bone. Thus, factors that inhibit osteoclast formation or bone resorption activity of mature osteoclasts may have the potential to inhibit the vicious cycle. The RANKL inhibitor denosumab is approved for treatment of bone metastases from solid tumors, and the cathepsin K inhibitor odanacatib has been shown to suppress bone resorption in breast cancer patients with bone metastases. Human osteoclasts can be generated from bone marrow-derived CD34+ mesenchymal stem cells in the presence of M-CSF and RANKL. In this study we report optimization of separate in vitro culture systems for determining osteoclast differentiation and activity, and validation of denosumab and odanacatib as reference inhibitors of osteoclast differentiation and activity, respectively. CD34+ human osteoclast precursor cells were cultured on bovine bone slices for 7 days. Different concentrations of denosumab (0.01 - 10 μg/ml) were added in the cultures at day 0, and tartrate-resistant acid phosphatase isoform 5b activity (TRACP 5b) was measured from the culture medium collected at day 7 as an index of the number of formed osteoclasts. Osteoclast activity was studied by allowing the formed mature osteoclasts to resorb bone during an additional 3-day culture period. The culture medium was changed and different concentrations of odanacatib (0.001 - 1.0 μM) were added into the cultures at day 7, and the amount of C-terminal cross-linked telopeptides of type I collagen (CTX-I) was measured in the culture medium collected at day 10 to quantitate bone resorption during days 7-10. Denosumab and odanacatib showed strong concentration dependent inhibition of osteoclast differentiation and activity, respectively, with EC50 values of 0.124 μg/ml for denosumab and 0.0433 μM for odanacatib. We conclude that we have validated denosumab as a reference compound of osteoclast differentiation and odanacatib as a reference compound of osteoclast activity in a human in vitro osteoclast culture system. The culture system is a clinically reliable tool for identifying new compounds affecting the vicious cycle of osteolytic bone metastases, and clarifying if these active compounds target directly osteoclast differentiation or activity. Citation Format: Jenni Bernoulli, Jussi M. Halleen, Mari I. Suominen, Johanna Tuomela, Jukka Rissanen, Katja M. Fagerlund. Validation of human osteoclast cultures for studying the mode-of-action and identification of new compounds with the potential of inhibiting the vicious cycle in osteolytic bone metastases. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 364.

Collaboration


Dive into the Katja M. Fagerlund's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Karl Ziegelbauer

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar

Arne Scholz

Bayer HealthCare Pharmaceuticals

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge