Network


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

Hotspot


Dive into the research topics where David Loven is active.

Publication


Featured researches published by David Loven.


Journal of Neuro-oncology | 2004

Non-resectable slow-growing meningiomas treated by hydroxyurea.

David Loven; Ruth Hardoff; Zvi Bar Sever; Adam Steinmetz; Michael Gornish; Zvi H. Rappaport; Eyal Fenig; Zvi Ram; Aaron Sulkes

AbstractPurpose: To test the benefit of hydroxyurea in the treatment of recurrent and non-resectable slow-growing meningiomas. Methods: Twelve patients with regrowing non-malignant meningiomas, were enrolled for a protocol of 2 years with continuous chemotherapy with hydroxyurea, 20 mg/kg/day. Response to treatment was evaluated both clinically and by diagnostic imaging using computed tomography (CT) and 201-Thallium single photon emission CT. One minimal response was documented by CT, accompanied by clinical stabilization. Nine patients showed progressive disease, at least by one imaging procedure, with a median time to progression of 13 months (range 4–24). Two other patients were not available for response due to early removal from the study, following abrupt manifestation of grades 3–4 hematological toxicity. Conclusion: In this series hydroxyurea has not shown effectiveness in the treatment of non-resectable slow-growing meningiomas: neither for achieving response, nor for arresting disease progression.


Cancer Research | 2011

Host Response to Short-term, Single-Agent Chemotherapy Induces Matrix Metalloproteinase-9 Expression and Accelerates Metastasis in Mice

Svetlana Gingis-Velitski; David Loven; Liat Benayoun; Michal Munster; Rotem Bril; Tali Voloshin; Dror Alishekevitz; Francesco Bertolini; Yuval Shaked

Mounting evidence suggests that bone marrow-derived cells (BMDC) contribute to tumor growth, angiogenesis, and metastasis. In acute reactions to cancer therapy, several types of BMDCs are rapidly mobilized to home tumors. Although this host reaction to therapy can promote tumor regrowth, its contribution to metastasis has not been explored. To focus only on the effects of chemotherapy on the host, we studied non-tumor-bearing mice. Plasma from animals treated with the chemotherapy paclitaxel induced angiogenesis, migration, and invasion of tumor cells along with host cell colonization. Lesser effects were seen with the chemotherapy gemcitabine. Conditioned medium from BMDCs and plasma from chemotherapy-treated mice each promoted metastatic properties in tumor cells by inducing matrix metalloproteinase-9 (MMP9) and epithelial-to-mesenchymal transition. In mice in which Lewis lung carcinoma cells were injected intravenously, treatment with paclitaxel, but not gemcitabine or vehicle, accelerated metastases in a manner that could be blocked by an MMP9 inhibitor. Moreover, chimeric mice reconstituted with BMDC where MMP9 activity was attenuated did not support accelerated metastasis by carcinoma cells that were pretreated with chemotherapy before their introduction to host animals. Taken together, our findings illustrate how some chemotherapies can exert prometastatic effects that may confound treatment outcomes.


Drug Discovery Today | 2013

Low-dose metronomic chemotherapy: from past experience to new paradigms in the treatment of cancer

David Loven; Erez Hasnis; Francesco Bertolini; Yuval Shaked

Low-dose metronomic (LDM) chemotherapy represents an emerging concept in the treatment of cancer. Directed against tumor cells and other types of cells, such as endothelial and immune cells, this treatment regimen alters the tumor microenvironment and suppresses innate features which support tumor growth. Ongoing Phase III clinical studies explore various applications of LDM chemotherapy, mostly combined with other anticancer agents, to act as complementary treatments to conventional maximum tolerated dose (MTD) chemotherapy. In this article we summarize preclinical and clinical experience with LDM chemotherapy, emphasizing the potential contribution of this new treatment modality to future paradigms in the systemic treatment of patients with cancer.


Neurosurgery | 1991

Radiation-induced cerebellar glioblastoma multiforme subsequent to treatment of an astrocytoma of the cervical spinal cord.

Zvi Harry Rappaport; David Loven; Uriel Ben-Aharon

A cerebellar glioblastoma multiforme was diagnosed in a 22-year-old woman. This originated in the zone adjacent to a field irradiated 14 years earlier after the removal of a noncontiguous astrocytoma of the spinal cord. The accepted criteria for radiation-induced tumors of the central nervous system are discussed.


Cell Reports | 2016

Macrophage-Induced Lymphangiogenesis and Metastasis following Paclitaxel Chemotherapy Is Regulated by VEGFR3

Dror Alishekevitz; Svetlana Gingis-Velitski; Orit Kaidar-Person; Lilach Gutter-Kapon; Sandra D. Scherer; Ziv Raviv; Emmanuelle Merquiol; Yael Ben-Nun; Valeria Miller; Chen Rachman-Tzemah; Michael Timaner; Yelena Mumblat; Neta Ilan; David Loven; Dov Hershkovitz; Ronit Satchi-Fainaro; Galia Blum; Jonathan P. Sleeman; Israel Vlodavsky; Yuval Shaked

Summary While chemotherapy strongly restricts or reverses tumor growth, the response of host tissue to therapy can counteract its anti-tumor activity by promoting tumor re-growth and/or metastases, thus limiting therapeutic efficacy. Here, we show that vascular endothelial growth factor receptor 3 (VEGFR3)-expressing macrophages infiltrating chemotherapy-treated tumors play a significant role in metastasis. They do so in part by inducing lymphangiogenesis as a result of cathepsin release, leading to VEGF-C upregulation by heparanase. We found that macrophages from chemotherapy-treated mice are sufficient to trigger lymphatic vessel activity and structure in naive tumors in a VEGFR3-dependent manner. Blocking VEGF-C/VEGFR3 axis inhibits the activity of chemotherapy-educated macrophages, leading to reduced lymphangiogenesis in treated tumors. Overall, our results suggest that disrupting the VEGF-C/VEGFR3 axis not only directly inhibits lymphangiogenesis but also blocks the pro-metastatic activity of macrophages in chemotherapy-treated mice.


International Journal of Cancer | 2014

Tumor‐derived microparticles induce bone marrow‐derived cell mobilization and tumor homing: A process regulated by osteopontin

Ella Fremder; Michal Munster; Anat Aharon; Valeria Miller; Svetlana Gingis-Velitski; Tali Voloshin; Dror Alishekevitz; Rotem Bril; Stefan J. Scherer; David Loven; Benjamin Brenner; Yuval Shaked

Acute chemotherapy can induce rapid bone‐marrow derived pro‐angiogenic cell (BMDC) mobilization and tumor homing, contributing to tumor regrowth. To study the contribution of tumor cells to tumor regrowth following therapy, we focused on tumor‐derived microparticles (TMPs). EMT/6 murine‐mammary carcinoma cells exposed to paclitaxel chemotherapy exhibited an increased number of TMPs and significantly altered their angiogenic properties. Similarly, breast cancer patients had increased levels of plasma MUC‐1+TMPs following chemotherapy. In addition, TMPs from cells exposed to paclitaxel induced higher BMDC mobilization and colonization, but had no increased effect on angiogenesis in Matrigel plugs and tumors than TMPs from untreated cells. Since TMPs abundantly express osteopontin, a protein known to participate in BMDC trafficking, the impact of osteopontin‐depleted TMPs on BMDC mobilization, colonization, and tumor angiogenesis was examined. Although EMT/6 tumors grown in mice inoculated with osteopontin‐depleted TMPs had lower numbers of BMDC infiltration and microvessel density when compared with EMT/6 tumors grown in mice inoculated with wild‐type TMPs, no significant difference in tumor growth was seen between the two groups. However, when BMDCs from paclitaxel‐treated mice were injected into wild‐type EMT/6‐bearing mice, a substantial increase in tumor growth and BMDC infiltration was detected compared to osteopontin‐depleted EMT/6‐bearing mice injected with BMDCs from paclitaxel‐treated mice. Collectively, our results suggest that osteopontin expressed by TMPs play an important role in BMDC mobilization and colonization of tumors, but is not sufficient to enhance the angiogenic activity in tumors.


Acta Oncologica | 2008

Daily low-dose/continuous capecitabine combined with neo-adjuvant irradiation reduces VEGF and PDGF-BB levels in rectal carcinoma patients

David Loven; Einat Beery; Rinat Yerushalmi; Claude Koren; Aaron Sulkes; Idit Lavi; Yuval Shaked; Eyal Fenig

Metronomic low-dose chemotherapy regimen was found to have an antiangiogenic effect in tumors. However, its effect on levels of circulating pro-angiogenic and anti-angiogenic factors is not fully explored. Materials and methods. The levels of both VEGF and PDGF-BB were measured in three time points, in the serum of 32 rectal carcinoma patients receiving daily reduced-dose/continuous capecitabine in combination with preoperative pelvic irradiation. Results. We found a significant decrease in VEGF and PDGF-BB serum levels during the combination treatment (p<0.0001), followed by an increase in the successive rest-period (p<0.0001). In addition, substantial changes in platelets counts were observed during treatment in correlation with the changes of VEGF and PDGF-BB serum levels. Discussion. These results suggest that combined chemo-irradiation affect levels of pro-angiogenic factors during treatment, and may reflect an anti-angiogenic window induced during this treatment. The potential implications of this inducible phenomenon, including a possible clinical benefit from the administration of long lasting metronomic chemotherapy immediately following combined chemo-irradiation, would warrant further investigation.


Molecular Cancer Therapeutics | 2014

Differential Therapeutic Effects of Anti–VEGF-A Antibody in Different Tumor Models: Implications for Choosing Appropriate Tumor Models for Drug Testing

Dror Alishekevitz; Rotem Bril; David Loven; Valeria Miller; Tali Voloshin; Svetlana Gingis-Velistki; Ella Fremder; Stefan J. Scherer; Yuval Shaked

We previously reported that the host response to certain chemotherapies can induce primary tumor regrowth, angiogenesis, and even metastases in mice, but the possible impact of anti–VEGF-A therapy in this context has not been fully explored. We, therefore, used combinations of anti–VEGF-A with chemotherapy on various tumor models in mice, including primary tumors, experimental lung metastases, and spontaneous lung metastases of 4T1-breast and CT26-colon murine cancer cell lines. Our results show that a combined treatment with anti–VEGF-A and folinic acid/5-fluorouracil/oxaliplatin (FOLFOX) but not with anti–VEGF-A and gemcitabine/cisplatinum (Gem/CDDP) enhances the treatment outcome partly due to reduced angiogenesis, in both primary tumors and experimental lung metastases models. However, neither treatment group exhibited an improved treatment outcome in the spontaneous lung metastases model, nor were changes in endothelial cell numbers found at metastatic sites. As chemotherapy has recently been shown to induce tumor cell invasion, we tested the invasion properties of tumor cells when exposed to plasma from FOLFOX-treated mice or patients with cancer. While plasma from FOLFOX-treated mice or patients induced invasion properties of tumor cells, the combination of anti–VEGF-A and FOLFOX abrogated these effects, despite the reduced plasma VEGF-A levels detected in FOLFOX-treated mice. These results suggest that the therapeutic impact of antiangiogenic drugs varies in different tumor models, and that anti–VEGF-A therapy can block the invasion properties of tumor cells in response to chemotherapy. These results may implicate an additional therapeutic role for anti–VEGF-A when combined with chemotherapy. Mol Cancer Ther; 13(1); 202–13. ©2013 AACR.


Oncotarget | 2017

Chemotherapy administration to breast cancer patients affects extracellular vesicles thrombogenicity and function

Anat Aharon; Anni Sabbah; Shahar Ben-Shaul; Hila Berkovich; David Loven; Benjamin Brenner; Gil Bar-Sela

Breast cancer (BC) is the most prevalent type of malignancy in women. Extracellular vesicles (EVs) are subcellular membrane blebs that include exosomes and microparticles. Study aims To elucidate the effects of chemotherapy administration on BC patients’ EVs characteristics and their effects on endothelial cells (EC) functions. Methods EVs were isolated from the blood samples of 54 BC patients treated by chemotherapy (25 neo-adjuvant, 29 adjuvant) and from 20 healthy women (control group). Blood samples were taken before chemotherapy and on the day of last chemotherapy administration. In some patients, samples were also evaluated 24 hours after chemotherapy treatment. EVs were characterized by cell origin, thrombogenicity and cytokine content. EVs effects on coagulation, migration, apoptosis and proliferation of endothelial cells were assessed as well. Results Patient characteristics of the two subgroups were similar except for tumor size. Change in EV expression of BC markers, MUC1 and EpCAM, were found in patient subgroups. EC-EVs were significantly higher in both patient subgroups compared to healthy controls. Higher EVs pro-coagulant activity was found at the end of chemotherapy and a significant increase in the ratio between tissue factor (TF) and TF pathway inhibitor was documented after the first 24hours of exposure to doxorubicin treatment. Furthermore, EVs of neo-adjuvant patients obtained before chemotherapy contained more pro-angiogenic proteins, reduced endothelial cells apoptosis and increased their migration compared to EVs obtained at the same timing from adjuvant patients. Conclusions EVs may serve as a biomarker for chemotherapy-related thrombogenicity and may indicate vascular damage even before chemotherapy.


Cancer | 1991

Soluble histocompatibility antigen class I in breast cancer patients in relation to tumor burden

Baruch Klein; Tirza Klein; Arie Figer; Margalit Bleiberg; Jermiahu Shapira; David Loven; Ella Livni; Hedwig Lurie; A. Niska

Serum beta‐2 microglobulin (B‐2M) levels were studied in 365 breast cancer patients and 210 age‐matched controls. The patients were divided into three groups: Group A, new patients at diagnosis; Group B, patients at follow‐up; and Group C, metastatic patients. The mean B‐2M of all breast cancer patients plus or minus one standard deviation (3.5 ± 1.2; range, 1.1 to 5.9) was significantly higher than normal controls (1.29 ± 0.49; range, 0.3 to 2.3; P < 0.005). When the three patient groups were compared with each other, the mean B‐2M level of Group A (3.0 ± 1.5; range, 0.9 to 6.9) was similar to that of Group C (4.22 ± 1.1; range, 2.0 to 6.4). The mean B‐2M of both Groups A and C was significantly higher than that of Group B (2.38 ± 1.02, range, 0.4 to 5.4; P < 0.001). In Group A the mean B‐2M decreased significantly after a 12‐month period and reached the mean level of Group B but not that of normal controls. When patients in Group B were analyzed by their stage of disease at diagnosis, there was no significant difference between Stages I and II. There was a significant difference in the mean B‐2M levels between Stages I and III. In relapsing patients, mean B‐2M levels increased. These findings suggest that serum B‐2M levels may reflect tumor burden, and even in patients at follow‐up, occult tumor cells may activate the immune system.

Collaboration


Dive into the David Loven's collaboration.

Top Co-Authors

Avatar

Yuval Shaked

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Dror Alishekevitz

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rotem Bril

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tali Voloshin

Technion – Israel Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Valeria Miller

Rappaport Faculty of Medicine

View shared research outputs
Top Co-Authors

Avatar

Anat Aharon

Rambam Health Care Campus

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge