Network


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

Hotspot


Dive into the research topics where Haifu Li is active.

Publication


Featured researches published by Haifu Li.


Onkologie | 2014

Soft tissue sarcoma: an update on systemic treatment options for patients with advanced disease.

Patrick Schöffski; Jasmien Cornillie; Agnieszka Wozniak; Haifu Li; Daphne Hompes

Sarcomas are a group of rare solid tumours arising from mesenchymal or connective tissue. This review focuses on soft tissue sarcoma and covers general topics such as the epidemiology, age distribution, site of disease, histogenesis, histological subtypes, prognosis and outcome of treatment. In more detail, the article reviews current systemic treatment standards and selected adverse events of agents such as doxorubicin, ifosfamide, trabectedin and pazopanib, and briefly highlights some drugs that are used off-label in specific subtypes of sarcoma.


Clinical Cancer Research | 2013

A potent combination of the novel PI3K inhibitor, GDC-0941, with imatinib in gastrointestinal stromal tumor xenografts: long-lasting responses after treatment withdrawal

Giuseppe Floris; Agnieszka Wozniak; Raf Sciot; Haifu Li; Lori S. Friedman; Thomas Van Looy; Jasmien Wellens; Peter Vermaelen; Christophe Deroose; Jonathan A. Fletcher; Maria Debiec-Rychter; Patrick Schöffski

PURPOSE Oncogenic signaling in gastrointestinal stromal tumors (GIST) is sustained via PI3K/AKT pathway. We used a panel of six GIST xenograft models to assess efficacy of GDC-0941 as single agent or in combination with imatinib (IMA). EXPERIMENTAL DESIGN Nude mice (n = 136) were grafted bilaterally with human GIST carrying diverse KIT mutations. Mice were orally dosed over four weeks, grouped as follows: (A) control; (B) GDC-0941; (C) imatinib, and (D) GDC+IMA treatments. Xenografts regrowth after treatment discontinuation was assessed in groups C and D for an additional four weeks. Tumor response was assessed by volume measurements, micro-PET imaging, histopathology, and immunoblotting. Moreover, genomic alterations in PTEN/PI3K/AKT pathway were evaluated. RESULTS In all models, GDC-0941 caused tumor growth stabilization, inhibiting tumor cell proliferation, but did not induce apoptosis. Under GDC+IMA, profound tumor regression, superior to either treatment alone, was observed. This effect was associated with the best histologic response, a nearly complete proliferation arrest and increased apoptosis. Tumor regrowth assays confirmed superior activity of GDC+IMA over imatinib; in three of six models, tumor volume remained reduced and stable even after treatment discontinuation. A positive correlation between response to GDC+IMA and PTEN loss, both on gene and protein levels, was found. CONCLUSION GDC+IMA has significant antitumor efficacy in GIST xenografts, inducing more substantial tumor regression, apoptosis, and durable effects than imatinib. Notably, after treatment withdrawal, tumor regression was sustained in tumors exposed to GDC+IMA, which was not observed under imatinib. Assessment of PTEN status may represent a useful predictive biomarker for patient selection.


Clinical Cancer Research | 2014

Phosphoinositide 3-Kinase Inhibitors Combined with Imatinib in Patient-Derived Xenograft Models of Gastrointestinal Stromal Tumors: Rationale and Efficacy

Thomas Van Looy; Agnieszka Wozniak; Giuseppe Floris; Raf Sciot; Haifu Li; Jasmien Wellens; Ulla Vanleeuw; Jonathan A. Fletcher; Paul W. Manley; Maria Debiec-Rychter; Patrick Schöffski

Introduction: The PI3K signaling pathway drives tumor cell proliferation and survival in gastrointestinal stromal tumor (GIST). We tested the in vivo efficacy of three PI3K inhibitors (PI3Ki) in patient-derived GIST xenograft models. Experimental Design: One hundred and sixty-eight nude mice were grafted with human GIST carrying diverse KIT genotypes and PTEN genomic status. Animals were dosed orally for two weeks as follows: control group (untreated); imatinib (IMA); PI3Ki (BKM120—buparlisib, BEZ235, or BYL719) or combinations of imatinib with a PI3Ki. Western blotting, histopathology, and tumor volume evolution were used for the assessment of treatment efficacy. Furthermore, tumor regrowth was evaluated for three weeks after treatment cessation. Results: PI3Ki monotherapy showed a significant antitumor effect, reflected in tumor volume reduction or stabilization, inhibitory effects on mitotic activity, and PI3K signaling inhibition. The IMA+PI3Ki combination remarkably improved the efficacy of either single-agent treatment with more pronounced tumor volume reduction and enhanced proapoptotic effects over either single agent. Response to IMA+PI3Ki was found to depend on the KIT genotype and specific model-related molecular characteristics. Conclusion: IMA+PI3Ki has significant antitumor efficacy in GIST xenografts as compared with single-agent treatment, resulting in more prominent tumor volume reduction and enhanced induction of apoptosis. Categorization of GIST based on KIT genotype and PI3K/PTEN genomic status combined with dose optimization is suggested for patient selection for clinical trials exploring such combinations. Clin Cancer Res; 20(23); 6071–82. ©2014 AACR.


Translational Oncology | 2014

Pazopanib, a Receptor Tyrosine Kinase Inhibitor, Suppresses Tumor Growth through Angiogenesis in Dedifferentiated Liposarcoma Xenograft Models

Haifu Li; Agnieszka Wozniak; Raf Sciot; Jasmien Cornillie; Jasmien Wellens; Thomas Van Looy; Ulla Vanleeuw; Marguerite Stas; Daphne Hompes; Maria Debiec-Rychter; Patrick Schöffski

INTRODUCTION: The rarity of dedifferentiated liposarcoma (DDLPS) and the lack of experimental DDLPS models limit the development of novel therapeutic strategies. Pazopanib (PAZ) is a tyrosine kinase inhibitor that is approved for the treatment of non-adipocytic advanced soft tissue sarcoma. The activity of this agent has not yet been properly explored in preclinical liposarcoma models nor in a randomized phase Ш clinical trial in this entity. The aim of the present study was to investigate whether PAZ had antitumor activity in DDLPS models in vivo. MATERIAL AND METHODS: We established two patient-derived DDLPS xenograft models (UZLX-STS3 and UZLX-STS5) through implantation of tumor material from sarcoma patients in athymic nude NMRI mice. An animal model of the SW872 liposarcoma cell line was also used. To investigate the efficacy of PAZ in vivo, mice bearing tumors were treated for 2 weeks with sterile water, doxorubicin (1.2 mg/kg, intraperitoneally, twice per week), PAZ [40 mg/kg, orally (p.o.), twice per day], or PAZ plus doxorubicin (same schedules as for single treatments). RESULTS: Patient-derived xenografts retained the histologic and molecular features of DDLPS. PAZ significantly delayed tumor growth by decreasing proliferation and inhibited angiogenesis in all models tested. Combining the angiogenesis inhibitor with an anthracycline did not show superior efficacy. CONCLUSION: These results suggest that PAZ has potential antitumor activity in DDLPS primarily through antiangiogenic effects and therefore should be explored in clinical trials.


Translational Oncology | 2015

Therapeutic Efficacy Assessment of CK6, a Monoclonal KIT Antibody, in a Panel of Gastrointestinal Stromal Tumor Xenograft Models

Thomas Van Looy; Agnieszka Wozniak; Giuseppe Floris; Haifu Li; Jasmien Wellens; Ulla Vanleeuw; Raf Sciot; Maria Debiec-Rychter; Patrick Schöffski

We evaluated the efficacy of CK6, a KIT monoclonal antibody, in a panel of human gastrointestinal stromal tumor (GIST) xenograft models. Nude mice were bilaterally transplanted with human GIST xenografts (four patient derived and two cell line derived), treated for 3 weeks, and grouped as follows: control (untreated); CK6 (40 mg/kg, 3 × weekly); imatinib (50 mg/kg, twice daily); sunitinib (40 mg/kg, once daily); imatinib + CK6; sunitinib + CK6 (same doses and schedules as in the single-agent treatments). Tumor volume assessment, Western blot analysis, and histopathology were used for evaluation of efficacy. Statistical analysis was performed using Mann-Whitney U (MWU) and Wilcoxon matched-pairs tests. CK6 as a single agent only reduced tumor growth rate in the UZLX-GIST3 model (P = .053, MWU compared to control), while in none of the other GIST models an effect on tumor growth rate was observed. CK6 did not result in significant anti-proliferative or pro-apoptotic effects in any of the GIST models, and moreover, CK6 did not induce a remarkable inhibition of KIT activation. Furthermore, no synergistic effect of combining CK6 with tyrosine kinase inhibitors (TKIs) was observed. Conversely, in certain GIST xenografts, anti-tumor effects seemed to be inferior under combination treatment compared to single-agent TKI treatment. In the GIST xenografts tested, the anti-tumor efficacy of CK6 was limited. No synergy was observed on combination of CK6 with TKIs in these GIST models. Our findings highlight the importance of using relevant in vivo human tumor xenograft models in the preclinical assessment of drug combination strategies.


Cancer Research | 2015

Abstract 1460: Establishment and characterization of a panel of patient-derived soft tissue sarcoma (STS) xenograft models for in vivo testing of novel therapeutic approaches

Haifu Li; Jasmien Cornillie; Agnieszka Wozniak; Thomas Van Looy; Yemarshet Kelemework Gebreyohannes; Jasmien Wellens; Ulla Vanleeuw; Daphne Hompes; Marguerite Stas; Friedl Sinnaeve; Maria Debiec-Rychter; Raf Sciot; Patrick Schöffski

Background: STS constitutes a rare and very heterogeneous family of tumors of mesenchymal origin. The limited treatment options available for advanced STS underline the need for reliable preclinical models to test novel therapeutic strategies. Methods: A panel of patient-derived xenografts was established and is currently expanded in nu/nu NMRI mice by subcutaneous implantation of fresh, surgically resected tumor specimens from consenting patients with STS, treated at the University Hospitals Leuven, Belgium. We mainly focused on STS subtypes which harbor specific genetic alterations (e.g. translocations, gene amplifications). Once tumor growth was observed, pieces of tissue were re-transplanted to the next generation of mice. At each passage tumor fragments were collected for histological and molecular characterization. Fluorescence in situ hybridization (FISH) was performed to confirm specific genomic alterations in selected xenografts. A STS xenograft model was considered established after observing maintained histological and molecular features for at least two passages. Results: Until now a total of 78 STS patient samples have been transplanted. Fifteen well-characterized, stable STS models have already been established, maintaining the features of the original tumor: myxofibrosarcoma (4 models), dedifferentiated liposarcoma (2), synovial sarcoma (2), epithelioid haemangioendothelioma (1), malignant peripheral nerve sheath tumor (2), mesenchymal chondrosarcoma (1), leiomyosarcoma (2) and sarcoma not otherwise specified (1). Some of these models have already been successfully used for in vivo testing of novel agents, including tyrosine kinase inhibitors or cytotoxic (pro-)drugs. Twelve other xenografts are still in early stages of engraftment, not yet fulfilling our criteria of “established model”. Conclusion: This newly established, representative panel of STS xenografts is characterized by stable histological and molecular features reflecting the pheno- and genotype of the original patient samples. This panel is thus well suited for in vivo drug testing of innovative agents. The unique availability of models of some ultra-rare entities will also allow us to study the biology of these diseases. The platform is available for collaboration with academic and commercial partners (contact [email protected]). Citation Format: Haifu Li, Jasmien Cornillie, Agnieszka Wozniak, Thomas Van Looy, Yemarshet Gebreyohannes, Jasmien Wellens, Ulla Vanleeuw, Daphne Hompes, Marguerite Stas, Friedl Sinnaeve, Maria Debiec-Rychter, Raf Sciot, Patrick Schoffski. Establishment and characterization of a panel of patient-derived soft tissue sarcoma (STS) xenograft models for in vivo testing of novel therapeutic approaches. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 1460. doi:10.1158/1538-7445.AM2015-1460


Cancer Research | 2015

Abstract 775: Anti-tumor effects of dovitinib, a multi-target kinase inhibitor, in patient-derived gastrointestinal stromal tumor (GIST) xenograft models

Yemarshet Kelemework Gebreyohannes; Thomas Van Looy; Agnieszka Wozniak; Jasmien Wellens; Haifu Li; Jasmien Cornillie; Ulla Vanleeuw; Lise Vreys; Matthew Squires; Ana-Maria Rodringuez; Maria Debiec-Rychter; Raf Sciot; Patrick Schöffski

Background: The majority of GISTs are driven by KIT/PDGFRA gain of function mutations. Advanced GIST is routinely treated with tyrosine kinase inhibitors (TKI; e.g imatinib, sunitinib and regorafenib). However, with time, heterogeneous resistance to these agents occurs, mainly due to acquired mutations in KIT. We tested the efficacy of dovitinib, which acts against VEGFR, FGFR, FLT3, PDGFRB and KIT, using patient-derived GIST xenograft models. Methods: NMRI nu/nu mice (n = 47) were bilaterally transplanted with the human GIST xenografts: a dose-dependent-imatinib-sensitive model UZLX-GIST2 (KIT: p.A502_Y503dup) or a model resistant to both imatinib and sunitinib, UZLX-GIST9 (KIT: p.P577del+p.W557LfsX5+p.D820G). Mice were divided to four treatment groups: control (untreated), imatinib (50 mg/kg/bid p.o.), imatinib (100 mg/kg/bid p.o.) and dovitinib (30 mg/kg/qd p.o.). Efficacy was assessed by tumor volume measurement (3x/week), histopathology [hematoxylin & eosin staining (HE p Conclusion: The multi-kinase inhibitor dovitinib showed anti-tumor efficacy in GIST xenograft models, though the effect was more pronounced in KIT exon 9 mutant (UZLX-GIST2). The decrease in MVD in both models suggested that the anti-tumor effect in these models was at least partially mediated by an anti-angiogenic effect of dovitinib. 1 Antonescu et al. Clin Cancer Res. 2005; 11:4182-90 Citation Format: Yemarshet Kelemework Gebreyohannes, Thomas Van Looy, Agnieszka Wozniak, Jasmien Wellens, Haifu Li, Jasmien Cornillie, Ulla Vanleeuw, Lise Vreys, Matthew Squires, Ana-Maria Rodringuez, Maria Debiec-Rychter, Raf Sciot, Patrick Schoffski. Anti-tumor effects of dovitinib, a multi-target kinase inhibitor, in patient-derived gastrointestinal stromal tumor (GIST) xenograft models. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 775. doi:10.1158/1538-7445.AM2015-775


Cancer Research | 2015

Abstract 5353: Efficacy of an innovative, enzyme-activated doxorubicin prodrug in patient-derived dedifferentiated liposarcoma and synovial sarcoma xenograft models

Jasmien Cornillie; Agnieszka Wozniak; Lise Vreys; Haifu Li; Thomas Van Looy; Jasmien Wellens; Ulla Vanleeuw; Andre Trouet; Peter Pokreisz; Daphne Hompes; Maria Debiec-Rychter; Raf Sciot; Patrick Schöffski

Background and objective: Dedifferentiated liposarcoma (DDLPS) and synovial sarcoma (SynSa) are aggressive malignant tumors of mesenchymal origin. Prognosis of advanced disease is poor with low response rates to first-line single agent doxorubicin (doxo) treatment, which is regarded as standard of care in metastatic disease. We evaluated the efficacy of PhAc-ALGP-doxorubicin (ALGP-doxo), a prodrug which is metabolized to doxo by peptidases present in the tumor microenvironment and/or tumor cells, in one SynSa and two DDLPS patient-derived xenografts. Methods: NMRI mice (n = 24) were transplanted subcutaneously on both flanks with human DDLPS (models UZLX-STS3 and UZLX-STS5) or SynSa (UZLX-STS7). Mice were randomized to three treatment groups: control (vehicle), doxo (0.03 mmol/kg for UZLX-STS3; 0.04 mmol/kg for UZLX-STS5 and UZLX-STS7) or ALGP-doxo (1.2 mmol/kg). Treatments were administered using intraperitoneal osmotic pumps, continuously releasing drugs for 7 days. Afterwards, half of mice from each group were sacrificed and tumors were collected. Remaining mice were monitored for another 14 days and tumors were collected on day 21. Treatment efficacy was assessed by tumor volume, hematoxylin and eosin (HE 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 5353. doi:10.1158/1538-7445.AM2015-5353


Cancer Research | 2016

Abstract 5197: Patient-derived xenograft (PDX) models of soft tissue sarcoma (STS): a preclinical platform for early drug testing

Agnieszka Wozniak; Jasmien Cornillie; Yemarshet Kelemework Gebreyohannes; Bram Boeckx; Haifu Li; Thomas Van Looy; Giuseppe Floris; Jasmien Wellens; Lise Vreys; Daphne Hompes; Marguerite Stas; Friedl Sinnaeve; Diether Lambrechts; Maria Debiec-Rychter; Raf Sciot; Patrick Schöffski

Background: STS constitutes a rare and very heterogeneous family of mesenchymal tumors. The limited treatment options available for advanced STS underline the need for reliable preclinical models to test novel therapeutic strategies. Methods: A panel of patient-derived xenografts (PDX) was established by subcutaneous implantation of fresh, surgically resected tumor specimens in immunodeficient athymic nude NMRI mice. Once tumor growth was observed, pieces of tumor were re-transplanted to next generations of mice. At each passage tumor fragments were collected for histolopathological and molecular characterization. A model was considered established after observing stable histological and molecular features for at least two passages. To evaluate the stability of the genomic profile we performed low-coverage whole-genome sequencing on DNA isolated from tumors obtained from at least two passages. In addition RNA-seq was used to better characterize the mutational profile of the ex-mouse tumors. Results: Until now 139 STS samples from consenting patients treated at the University Hospitals, Leuven, Belgium, have been transplanted. Twenty six well-characterized, stable STS PDX models have been established, maintaining the histopathological and molecular features of the original tumor. Seventeen models, analyzed with low-coverage DNA sequencing, showed a stable genomic profile in at least two different tumor passages. At this point the panel includes models of gastrointestinal stromal tumor (6 models), myxofibrosarcoma (5), dedifferentiated liposarcoma (3), malignant peripheral nerve sheath tumor (3), synovial sarcoma (2), leiomyosarcoma (3) epithelioid haemangioendothelioma (1), mesenchymal chondrosarcoma (1) and undifferentiated high grade sarcoma (2). Some of these models have already been successfully used for in vivo testing of novel agents, including both targeted and cytotoxic (pro-)drugs, and results served as a rationale for at least four prospective clinical trials. In addition 22 other xenografts are still in early stages of engraftment, not yet fulfilling our criteria of an “established model”. Conclusion: Our panel of mesenchymal PDX models is characterized by stable histological and molecular features. These clinically well-annotated models can contribute to reliable preclinical studies for new anticancer treatments for STS. The availability of unique, rare STS xenografts also allows to study the biology of these diseases. The platform is made available to collaborators from academia and industry. Citation Format: Agnieszka Wozniak, Jasmien Cornillie, Yemarshet K. Gebreyohannes, Bram Boeckx, Haifu Li, Thomas Van Looy, Giuseppe Floris, Jasmien Wellens, Lise Vreys, Daphne Hompes, Marguerite Stas, Friedl Sinnaeve, Diether Lambrechts, Maria Debiec-Rychter, Raf Sciot, Patrick Schoffski. Patient-derived xenograft (PDX) models of soft tissue sarcoma (STS): a preclinical platform for early drug testing. [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 5197.


Cancer Research | 2015

Abstract 774: Cabozantinib, a multi-target receptor tyrosine kinase inhibitor, decreases tumor growth and angiogenesis in a patient-derived dedifferentiated liposarcoma xenograft

Haifu Li; Agnieszka Wozniak; Karel Van den Bossche; Thomas Van Looy; Jasmien Wellens; Daphne Hompes; Dana T. Aftab; Maria Debiec-Rychter; Raf Sciot; Patrick Schöffski

Proceedings: AACR 106th Annual Meeting 2015; April 18-22, 2015; Philadelphia, PA Background: Upregulation of receptor tyrosine kinase signaling pathways, including vascular endothelial growth factor receptor (VEGFR) and MET (also known as hepatocyte growth factor receptor), is found in different subtypes of soft tissue sarcoma. Cabozantinib (CABO) is a multi-target inhibitor of receptor tyrosine kinases including VEGFR, MET, RET, and KIT. We aimed to evaluate the anti-angiogenic and anti-tumor effect of CABO alone and in combination with the standard-of-care cytotoxic agent doxorubicin (DOX) in a patient-derived xenograft model of dedifferentiated liposarcoma (DDLPS). Methods: NMRI nu/nu mice were bilaterally transplanted with human UZLX-STS3 DDLPS tumors, a model originating from a patient with DDLPS. Twenty eight tumor-bearing mice were assigned to 4 groups and treated for three weeks with either vehicle (control), DOX (i.p., 1.2mg/kg/twice per week), CABO (p.o., 30mg/kg/QD) or CABO+DOX (same dose/schedule as for the single agents). Tumor volume was measured three times a week. Mitotic and apoptotic activity were assessed on hematoxylin and eosin (H&E) staining and with phospho-histone H3 (pHH3) and cleaved-caspase 3 (CC3) immunostainings. Microvascular density (MVD) was evaluated by counting CD34 positive vessels. Statistics were calculated using the Mann-Whitney U pairs tests. p < 0.05 was considered as statistically significant. Results: A significant delay in tumor growth was observed in CABO and CABO+DOX treated groups as compared to controls (p<0.05) or DOX single agent (p<0.05). Both CABO and CABO+DOX reduced the mitotic activity in comparison with control (38% and 66% decrease, respectively, p<0.005). CABO+DOX showed better antimitotic activity than either single agent DOX or CABO (p<0.005 for both). The CABO-based treatments induced higher apoptotic activity as compared to control (2.1 fold in CABO and 2.4 fold in CABO+DOX, p<0.005). Results were confirmed by pHH3 and CC3 stainings. Furthermore, significantly decreased MVD was observed in the CABO and the combination treated tumors as compared to control (64% and 67% decrease, respectively, p<0.005) and as compared to DOX alone (57% and 61% decrease, respectively, p<0.005). Conclusions: CABO has anti-tumor activity in the UZLX-STS3 DDLPS xenograft model by inhibiting proliferation, inducing apoptosis and suppressing angiogenesis. These in vivo results support ongoing and planned clinical trials with cabozantinib in mesenchymal malignancies. Citation Format: Haifu Li, Agnieszka Wozniak, Karel Van Den Bossche, Thomas Van Looy, Jasmien Wellens, Daphne Hompes, Dana Aftab, Maria Debiec-Rychter, Raf Sciot, Patrick Schoffski. Cabozantinib, a multi-target receptor tyrosine kinase inhibitor, decreases tumor growth and angiogenesis in a patient-derived dedifferentiated liposarcoma xenograft. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 774. doi:10.1158/1538-7445.AM2015-774

Collaboration


Dive into the Haifu Li's collaboration.

Top Co-Authors

Avatar

Agnieszka Wozniak

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Jasmien Wellens

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Raf Sciot

Catholic University of Leuven

View shared research outputs
Top Co-Authors

Avatar

Thomas Van Looy

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Jasmien Cornillie

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Daphne Hompes

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maria Debiec-Rychter

The Catholic University of America

View shared research outputs
Top Co-Authors

Avatar

Ulla Vanleeuw

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge