Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
Eur J Haematol ; 111(6): 938-945, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37731314

ABSTRACT

OBJECTIVES: Acquired missense mutations in the BCR::ABL1 kinase domain (KD) may cause tyrosine kinase inhibitor (TKI) treatment failure. Based on mutation-specific in vitro derived IC50-values, alternative TKI may be selected. We assessed clinical practice of BCR::ABL1 KD mutation testing, clinical response in relation to IC50-values, and clinical outcome of tested patients. METHODS: Patients from six Dutch CML reference centers and a national registry were included once a mutational analysis was performed. Reasons for testing were categorized as suboptimal TKI response, and primary or secondary TKI resistance. RESULTS: Four hundred twenty analyses were performed in 275 patients. Sixty-nine patients harbored at least one mutation. Most analyses were performed because of suboptimal TKI response but with low mutation incidence (4%), while most mutations were found in primary and secondary resistant patients (21% and 51%, respectively). Harboring a BCR::ABL1 mutation was associated with inferior overall survival (HR 3.2 [95% CI, 1.7-6.1; p < .001]). Clinically observed responses to TKI usually corresponded with the predicted TKI sensitivity based on the IC50-values, but a high IC50-value did not preclude a good clinical response per se. CONCLUSIONS: We recommend BCR::ABL1 KD mutation testing in particular in the context of primary or secondary resistance. IC50-values can direct the TKI choice for CML patients, but clinical efficacy can be seen despite adverse in vitro resistance.


Subject(s)
Fusion Proteins, bcr-abl , Leukemia, Myelogenous, Chronic, BCR-ABL Positive , Humans , Fusion Proteins, bcr-abl/genetics , Drug Resistance, Neoplasm/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mutation , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology
4.
Clin Microbiol Infect ; 29(4): 538.e1-538.e6, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36509372

ABSTRACT

OBJECTIVES: We report a patient case of pseudomembranous colitis associated with a monotoxin-producing Clostridioides difficile belonging to the very rarely diagnosed polymerase chain reaction (PCR) ribotype (RT) 151. To understand why this isolate was not identified using a routine commercial test, we performed a genomic analysis of RT151. METHODS: Illumina short-read sequencing was performed on n = 11 RT151s from various geographical regions to study their genomic characteristics and relatedness. Subsequently, we used PacBio circular consensus sequencing to determine the complete genome sequence of isolates belonging to cryptic clades C-I and C-II, which includes the patient isolate. RESULTS: We found that 1) RT151s are polyphyletic with isolates falling into clades 1 and cryptic clades C-I and C-II; 2) RT151 contains both nontoxigenic and toxigenic isolates and 3) RT151 C-II isolates contained monotoxin pathogenicity loci. The isolate from our patient case report contains a novel-pathogenicity loci insertion site, lacked tcdA and had a divergent tcdB sequence that might explain the failure of the diagnostic test. DISCUSSION: This study shows that RT151 encompasses both typical and cryptic clades and provides conclusive evidence for C. difficile infection due to clade C-II isolates that was hitherto lacking. Vigilance towards C. difficile infection as a result of cryptic clade isolates is warranted.


Subject(s)
Bacterial Toxins , Clostridioides difficile , Clostridium Infections , Humans , Bacterial Toxins/genetics , Ribotyping , Clostridium Infections/diagnosis , Polymerase Chain Reaction , Genomics
7.
Hemasphere ; 5(6): e580, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34095763

ABSTRACT

Various allogeneic (allo) stem cell transplantation platforms have been developed over the last 2 decades. In this review we focus on the impact of in vivo and ex vivo graft manipulation on immune reconstitution and clinical outcome. Strategies include anti-thymocyte globulin- and post-transplantation cyclophosphamide-based regimens, as well as graft engineering, such as CD34 selection and CD19/αßT cell depletion. Differences in duration of immune suppression, reconstituting immune repertoires, and associated graft-versus-leukemia effects and toxicities mediated through viral reactivations are highlighted. In addition, we discuss the impact of different reconstituting repertoires on donor lymphocyte infusions and post allo pharmacological interventions to enhance tumor control. We advocate for precisely counting all graft ingredients and therapeutic drug monitoring during conditioning in the peripheral blood, and for adjusting dosing accordingly on an individual basis. In addition, we propose novel trial designs to better assess the impact of variations in transplantation platforms in order to better learn from our diversity of "counts" and potential "adjustments." This will, in the future, allow daily clinical practice, strategic choices, and future trial designs to be based on data guided decisions, rather than relying on dogma and habits.

8.
Front Physiol ; 12: 638027, 2021.
Article in English | MEDLINE | ID: mdl-33708142

ABSTRACT

Familial hypobetalipoproteinemia is a metabolic disorder mainly caused by mutations in the apolipoprotein B gene. In its homozygous form it can lead without treatment to severe ophthalmological and neurological manifestations. In contrast, the heterozygous form is generally asymptomatic but associated with a low risk of cardiovascular disease. Acanthocytes or thorny red blood cells (RBCs) are described for both forms of the disease. However, those morphological changes are poorly characterized and their potential consequences for RBC functionality are not understood. Thus, in the present study, we asked whether, to what extent and how acanthocytes from a patient with heterozygous familial hypobetalipoproteinemia could exhibit altered RBC functionality. Acanthocytes represented 50% of the total RBC population and contained mitoTracker-positive surface patches, indicating the presence of mitochondrial fragments. While RBC osmotic fragility, calcium content and ATP homeostasis were preserved, a slight decrease of RBC deformability combined with an increase of intracellular free reactive oxygen species were observed. The spectrin cytoskeleton was altered, showing a lower density and an enrichment in patches. At the membrane level, no obvious modification of the RBC membrane fatty acids nor of the cholesterol content were detected but the ceramide species were all increased. Membrane stiffness and curvature were also increased whereas transversal asymmetry was preserved. In contrast, lateral asymmetry was highly impaired showing: (i) increased abundance and decreased functionality of sphingomyelin-enriched domains; (ii) cholesterol enrichment in spicules; and (iii) ceramide enrichment in patches. We propose that oxidative stress induces cytoskeletal alterations, leading to increased membrane stiffness and curvature and impaired lipid lateral distribution in domains and spicules. In addition, ceramide- and spectrin-enriched patches could result from a RBC maturation defect. Altogether, the data indicate that acanthocytes are associated with cytoskeletal and membrane lipid lateral asymmetry alterations, while deformability is only mildly impaired. In addition, familial hypobetalipoproteinemia might also affect RBC precursors leading to disturbed RBC maturation. This study paves the way for the potential use of membrane biophysics and lipid vital imaging as new methods for diagnosis of RBC disorders.

9.
Cancer Chemother Pharmacol ; 81(5): 911-921, 2018 05.
Article in English | MEDLINE | ID: mdl-29574584

ABSTRACT

PURPOSE: Chemotherapy-resistance remains a major obstacle to effective anti-cancer treatment. We previously showed that platinum analogs cause the release of two fatty acids. These platinum-induced fatty acids (PIFAs) induced complete chemoresistance in mice, whereas co-administration of a COX-1 inhibitor, indomethacin, prevented PIFA release and significantly enhanced chemosensitivity. To assess the safety of combining indomethacin with platinum-based chemotherapy, and to explore its efficacy and associated PIFA levels, a multi-center phase I trial was conducted. METHODS: The study was comprised of two arms: oxaliplatin plus capecitabine (CAPOX, arm I) and cisplatin plus gemcitabine, capecitabine or 5FU (arm II) in patients for whom these regimens were indicated as standard care. Indomethacin was escalated from 25 to 75 mg TID, using a standard 3 × 3 design per arm, and was administered orally 8 days around chemo-infusion from cycle two onwards. PIFA levels were measured before and after treatment initiation, with and without indomethacin. RESULTS: Thirteen patients were enrolled, of which ten were evaluable for safety analyses. In arm I, no dose-limiting toxicities were observed, and all indomethacin dose levels were well-tolerated. Partial responses were observed in three patients (30%). Indomethacin lowered plasma levels of 12-S-hydroxy-5,8,10-heptadecatrienoic acid (12-S-HHT), whereas 4,7,10,13-hexadecatetraenoic acid (16:4(n-3)) levels were not affected. Only one patient was included in arm II; renal toxicity led to closure of this cohort. CONCLUSIONS: Combined indomethacin and CAPOX treatment is safe and reduces the concentrations of 12-S-HHT, which may be associated with improved chemosensitivity. The recommended phase II dose is 75 mg indomethacin TID given 8 days surrounding standard dosed CAPOX.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Indomethacin/pharmacology , Neoplasms/drug therapy , Organoplatinum Compounds/pharmacology , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cyclooxygenase 1/metabolism , Cyclooxygenase Inhibitors/therapeutic use , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Resistance, Neoplasm/drug effects , Fatty Acids/metabolism , Female , Humans , Indomethacin/therapeutic use , Male , Middle Aged , Neoplasms/pathology , Organoplatinum Compounds/therapeutic use , Prospective Studies , Treatment Outcome
10.
FASEB J ; 31(5): 2195-2209, 2017 05.
Article in English | MEDLINE | ID: mdl-28183801

ABSTRACT

Although chemotherapy is designed to eradicate tumor cells, it also has significant effects on normal tissues. The platinum-induced fatty acid 16:4(n-3) (hexadeca-4,7,10,13-tetraenoic acid) induces systemic resistance to a broad range of DNA-damaging chemotherapeutics. We show that 16:4(n-3) exerts its effect by activating splenic F4/80+/CD11blow macrophages, which results in production of chemoprotective lysophosphatidylcholines (LPCs). Pharmacologic studies, together with analysis of expression patterns, identified GPR120 on F4/80+/CD11blow macrophages as the relevant receptor for 16:4(n-3). Studies that used splenocytes from GPR120-deficient mice have confirmed this conclusion. Activation of the 16:4(n-3)-GPR120 axis led to enhanced cPLA2 activity in these splenic macrophages and secretion of the resistance-inducing lipid mediator, lysophosphatidylcholine(24:1). These studies identify a novel and unexpected function for GPR120 and suggest that antagonists of this receptor might be effective agents to limit development of chemotherapy resistance.-Houthuijzen, J. M., Oosterom, I., Hudson, B. D., Hirasawa, A., Daenen, L. G. M., McLean, C. M., Hansen, S. V. F., van Jaarsveld, M. T. M., Peeper, D. S., Jafari Sadatmand, S., Roodhart, J. M. L., van de Lest, C. H. A., Ulven, T., Ishihara, K., Milligan, G., Voest, E. E. Fatty acid 16:4(n-3) stimulates a GPR120-induced signaling cascade in splenic macrophages to promote chemotherapy resistance.


Subject(s)
Macrophages/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Animals , Drug Resistance/physiology , Fatty Acids, Omega-3/metabolism , Mice, Inbred BALB C , Signal Transduction/physiology
12.
JAMA Oncol ; 1(3): 350-8, 2015 Jun.
Article in English | MEDLINE | ID: mdl-26181186

ABSTRACT

IMPORTANCE: Our research group previously identified specific endogenous platinum-induced fatty acids (PIFAs) that, in picomolar quantities, activate splenic macrophages leading to resistance to chemotherapy in mouse models. Fish oil was shown to contain the PIFA 16:4(n-3) (hexadeca-4,7,10,13-tetraenoic acid) and when administered to mice neutralized chemotherapy activity. OBJECTIVE: Because patients with cancer frequently use fish oil supplements, we set out to determine exposure to 16:4(n-3) after intake of fish or fish oil. DESIGN, SETTING, AND PARTICIPANTS: (1) In November 2011, 400 patients with cancer undergoing treatment at the University Medical Center Utrecht were surveyed to determine their use of fish oil supplements; 118 patients responded to the questionnaire (30%); (2) pharmacokinetic analysis of the 16:4(n-3) content of 6 fish oils and 4 fishes was carried out; (3) from April through November 2012, a healthy volunteer study was performed to determine 16:4(n-3) plasma levels after intake of 3 different brands of fish oil or 4 different fish species. Thirty healthy volunteers were randomly selected for the fish oil study; 20 were randomly selected for the fish study. These studies were supported by preclinical tumor experiments in mice to determine chemoresistance conducted between September 2011 and December 2012. MAIN OUTCOMES AND MEASURES: (1) Rate of use of fish oil supplements among patients undergoing cancer treatment at our institution; (2) levels of 16:4(n-3) present in 3 brands of fish oil and 4 species of fish; and (3) plasma levels of 16:4(n-3) present in healthy volunteers after consuming fish oil or fish. RESULTS: Eleven percent of respondents reported using omega-3 supplements. All fish oils tested contained relevant amounts of 16:4(n-3), from 0.2 to 5.7 µM. Mouse experiments showed that addition of 1 µL of fish oil to cisplatin was sufficient to induce chemoresistance, treatment having no impact on the growth rate of tumors compared with vehicle-treated controls (estimated tumor volume difference, 44.1 mm3; P > .99). When the recommended daily amount of 10 mL of fish oil was administered to healthy volunteers, rises in plasma 16:4(n-3) levels were observed, reaching up to 20 times the baseline levels. Herring and mackerel contained high levels of 16:4(n-3) in contrast to salmon and tuna. Consumption of fish with high levels of 16:4(n-3) also resulted in elevated plasma levels of 16:4(n-3). CONCLUSIONS AND RELEVANCE: All tested fish oils and herring and mackerel fishes contained relevant levels of fatty acid 16:4(n-3), a fatty acid with chemotherapy-negating effects in preclinical models. After ingestion of these fish oils or fishes, 16:4(n-3) was rapidly taken up in the plasma of human volunteers. Until further data become available, fish oil and fish containing high levels of 16:4(n-3) may best be avoided on the days surrounding chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Dietary Supplements , Drug Resistance, Neoplasm , Fatty Acids/blood , Fish Oils/administration & dosage , Fishes , Food-Drug Interactions , Neoplasms/drug therapy , Seafood , Academic Medical Centers , Animals , Biomarkers/blood , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Line, Tumor , Cisplatin/pharmacology , Dose-Response Relationship, Drug , Fish Oils/pharmacokinetics , Health Care Surveys , Humans , Irinotecan , Mice, Inbred BALB C , Neoplasms/blood , Neoplasms/pathology , Netherlands , Organoplatinum Compounds/pharmacology , Oxaliplatin , Surveys and Questionnaires , Tumor Burden , Up-Regulation
13.
Nat Commun ; 5: 5275, 2014 Nov 12.
Article in English | MEDLINE | ID: mdl-25387467

ABSTRACT

Host responses to systemic anti-cancer treatment play important roles in the development of anti-cancer drug resistance. Here we show that F4/80(+)/CD11b(low) splenocytes mediate the resistance to DNA-damaging chemotherapeutics induced by two platinum-induced fatty acids (PIFAs), 12-S-keto-5,8,10-heptadecatrienoic acid and 4,7,10,13-hexadecatetraenoic acid (16:4(n-3)) in xenograft mouse models. Splenectomy or depletion of splenic macrophages by liposomal clodronate protects against PIFA-induced chemoresistance. In addition, we find that 12-S-HHT, but not 16:4(n-3), functions via leukotriene B4 receptor 2 (BLT2). Genetic loss or chemical inhibition of BLT2 prevents 12-S-HHT-mediated resistance. Mass spectrometry analysis of conditioned medium derived from PIFA-stimulated splenic macrophages identifies several lysophosphatidylcholines as the resistance-inducing molecules. When comparing cisplatin and PIFA-treated tumours with cisplatin alone treated tumours we found overall less γH2AX, a measure for DNA damage. Taken together, we have identified an intricate network of lysophospholipid signalling by splenic macrophages that induces systemic chemoresistance in vivo via an altered DNA damage response.


Subject(s)
DNA Damage , Drug Resistance, Neoplasm/physiology , Lysophospholipids/physiology , Macrophages/metabolism , Animals , Antineoplastic Agents/therapeutic use , Cisplatin/therapeutic use , DNA Damage/physiology , Fatty Acids, Unsaturated/metabolism , Fatty Acids, Unsaturated/physiology , Lysophospholipids/metabolism , Macrophages/physiology , Male , Mass Spectrometry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms, Experimental/drug therapy , Receptors, Leukotriene B4/physiology , Spleen/cytology , Splenectomy
14.
J Pathol ; 231(1): 117-29, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23744542

ABSTRACT

Biallelic mutations of the von Hippel-Lindau (VHL) gene are the most common cause of sporadic and inherited renal cell carcinoma (RCC). Loss of VHL has been reported to affect cell proliferation by deregulating cell cycle-associated proteins. We report that the VHL gene product (pVHL) inhibits E2F1 expression at both mRNA and protein level in zebrafish and human RCC cells, while loss of VHL increases E2F1 expression in patient kidney tumour tissue and RCC cells, resulting in a delay of cell cycle progression. RCCs from von Hippel-Lindau patients with known germline VHL mutations express significantly more E2F1 compared to sporadic RCCs with either clear-cell (cc) or non-cc histology. Analysis of 138 primary RCCs reveals that E2F1 expression is significantly higher in tumours with a diameter ≤7 cm and with a favourable American Joint Committee on Cancer (AJCC) stage. The expression of E2F1 in RCC significantly correlates with p27 expression, suggesting that increased expression of E2F1 in RCC induces tumour cell senescence via p27. Cox regression analysis shows significant prediction of E2F1 expression for disease-free survival and overall survival, implying that E2F1 expression in kidney tumour is a novel prognostic factor for patients with RCC.


Subject(s)
Carcinoma, Renal Cell/mortality , E2F1 Transcription Factor/genetics , Gene Expression Regulation, Neoplastic/physiology , Kidney Neoplasms/mortality , Von Hippel-Lindau Tumor Suppressor Protein/physiology , Animals , Blotting, Western , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Cellular Senescence , Disease Models, Animal , E2F1 Transcription Factor/metabolism , Female , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Middle Aged , Organisms, Genetically Modified , Plasmids , Prognosis , Proliferating Cell Nuclear Antigen/metabolism , Proportional Hazards Models , Real-Time Polymerase Chain Reaction , Survival Rate , Transfection , Tumor Cells, Cultured , Zebrafish
15.
Blood ; 122(1): 143-53, 2013 Jul 04.
Article in English | MEDLINE | ID: mdl-23690447

ABSTRACT

Host responses to chemotherapy can induce resistance mechanisms that facilitate tumor regrowth. To determine the contribution of bone marrow-derived cells (BMDCs), we exposed tumor-bearing mice to chemotherapeutic agents and evaluated the influx and contribution of a genetically traceable subpopulation of BMDCs (vascular endothelial-cadherin-Cre-enhanced yellow fluorescent protein [VE-Cad-Cre-EYFP]). Treatment of tumor-bearing mice with different chemotherapeutics resulted in a three- to 10-fold increase in the influx of VE-Cad-Cre-EYFP. This enhanced influx was accompanied by a significant increase in angiogenesis. Expression profile analysis revealed a progressive change in the EYFP population with loss of endothelial markers and an increase in mononuclear markers. In the tumor, 2 specific populations of VE-Cad-Cre-EYFP BMDCs were identified: Gr1⁺/CD11b⁺ and Tie2high/platelet endothelial cell adhesion moleculelow cells, both located in perivascular areas. A common signature of the EYFP population that exits the bone marrow is an increase in Notch. Inducible inactivation of Notch in the EYFP⁺ BMDCs impaired homing of these BMDCs to the tumor. Importantly, Notch deletion reduced therapy-enhanced angiogenesis, and was associated with an increased antitumor effect of the chemotherapy. These findings revealed the functional significance of a specific population of supportive BMDCs in response to chemotherapeutics and uncovered a new potential strategy to enhance anticancer therapy.


Subject(s)
Bone Marrow Cells/physiology , Carcinoma, Lewis Lung/drug therapy , Cisplatin/pharmacology , Colorectal Neoplasms/drug therapy , Paclitaxel/pharmacology , Receptor, Notch1/physiology , Animals , Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Bone Marrow Cells/cytology , Cadherins/metabolism , Carcinoma, Lewis Lung/genetics , Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm/physiology , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/genetics , Mice , Mice, 129 Strain , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Receptor, Notch1/genetics , Xenograft Model Antitumor Assays
16.
J Urol ; 188(2): 607-14, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22704448

ABSTRACT

PURPOSE: Immune based therapy has gained renewed interest in the search for treatment strategies for metastasized renal cell carcinoma. We determined whether radio frequency ablation combined with interleukin-2 would induce a tumor specific immune response and serve as an in situ vaccine against renal cell carcinoma. MATERIALS AND METHODS: Mice with orthotopic renal tumors were treated with radio frequency ablation combined with interleukin-2, radio frequency ablation only, interleukin-2 only or no treatment as the control. Immunohistochemistry was done to determine which cells were present in the tumor after treatment. In vitro tumor specific cytotoxicity assays were performed with CD8+ T and natural killer cells derived from the spleen of treated mice. To study whether treatment could prevent metastasis or affect the growth of established metastases we induced lung metastasis intravenously before or after treatment and subsequently quantified it. RESULTS: The number of natural killer, CD4+ and CD8+ T cells was significantly increased in the tumor tissue of radio frequency ablation/interleukin-2 treated mice (p <0.0001). Natural killer and CD8+ T cells showed strong antitumor activity in vitro after combination treatment. Lung metastatic formation was significantly prevented in mice that received combination therapy (p <0.0001). Lung metastases were significantly smaller after combination treatment (vs interleukin-2 p = 0.025). CONCLUSIONS: Radio frequency ablation of the primary tumor combined with interleukin-2 induces a systemic antitumor immune response to renal cell carcinoma, which is much stronger than that of interleukin-2 monotherapy. This effect appears to be mediated by natural killer and CD8+ T cells. It may have an important role in the development of new immunotherapy strategies for renal cell carcinoma.


Subject(s)
Antineoplastic Agents/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/therapy , Catheter Ablation , Disease Models, Animal , Immunotherapy, Active/methods , Interleukin-2/administration & dosage , Kidney Neoplasms/immunology , Kidney Neoplasms/therapy , Killer Cells, Natural/immunology , Animals , Carcinoma, Renal Cell/secondary , Cell Line, Tumor , Combined Modality Therapy , Cytotoxicity Tests, Immunologic , Disease Progression , Humans , Lung Neoplasms/immunology , Lung Neoplasms/secondary , Lung Neoplasms/therapy , Male , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Spleen/immunology
17.
Cancer Res ; 71(22): 6976-85, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21975929

ABSTRACT

Recent studies suggest that chemotherapy, in addition to its cytotoxic effects on tumor cells, can induce a cascade of host events to support tumor growth and spread. Here, we used an experimental pulmonary metastasis model to investigate the role of this host response in metastasis formation. Mice were pretreated with chemotherapy and after clearance of the drugs from circulation, tumor cells were administered intravenously to study potential "protumorigenic" host effects of chemotherapy. Pretreatment with the commonly used chemotherapeutic agents cisplatin and paclitaxel significantly enhanced lung metastasis in this model. This corresponded to enhanced adhesion of tumor cells to an endothelial cell monolayer that had been pretreated with chemotherapy in vitro. Interestingly, chemotherapy exposure enhanced the expression of VEGF receptor 1 (VEGFR-1) on endothelial cells both in vitro and in vivo. Administration of antibodies targeting VEGFR-1 reversed the early retention of tumor cells in the lungs, thereby preventing the formation of chemotherapy-induced pulmonary metastases. The data indicate that chemotherapy-induced expression of VEGFR-1 on endothelial cells can create an environment favorable to tumor cell homing. Inhibition of VEGFR-1 function may therefore be used to counteract chemotherapy-induced retention of tumor cells within the metastatic niche, providing a novel level of tumor control in chemotherapy.


Subject(s)
Endothelial Cells/physiology , Neoplasms, Experimental/drug therapy , Vascular Endothelial Growth Factor Receptor-1/physiology , Animals , COS Cells , Cell Adhesion , Chlorocebus aethiops , Lung Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms, Experimental/pathology , Platelet Endothelial Cell Adhesion Molecule-1/analysis , Vascular Endothelial Growth Factor Receptor-1/analysis , Vascular Endothelial Growth Factor Receptor-1/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
18.
Cancer Cell ; 20(3): 370-83, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21907927

ABSTRACT

The development of resistance to chemotherapy is a major obstacle for lasting effective treatment of cancer. Here, we demonstrate that endogenous mesenchymal stem cells (MSCs) become activated during treatment with platinum analogs and secrete factors that protect tumor cells against a range of chemotherapeutics. Through a metabolomics approach, we identified two distinct platinum-induced polyunsaturated fatty acids (PIFAs), 12-oxo-5,8,10-heptadecatrienoic acid (KHT) and hexadeca-4,7,10,13-tetraenoic acid (16:4(n-3)), that in minute quantities induce resistance to a broad spectrum of chemotherapeutic agents. Interestingly, blocking central enzymes involved in the production of these PIFAs (cyclooxygenase-1 and thromboxane synthase) prevents MSC-induced resistance. Our findings show that MSCs are potent mediators of resistance to chemotherapy and reveal targets to enhance chemotherapy efficacy in patients.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclooxygenase 1/metabolism , Drug Resistance, Neoplasm , Fatty Acids, Unsaturated/metabolism , Fatty Acids/metabolism , Mesenchymal Stem Cells/metabolism , Platinum Compounds/pharmacology , Thromboxane-A Synthase/metabolism , Animals , Apoptosis/drug effects , Carboplatin/administration & dosage , Carboplatin/pharmacology , Cisplatin/administration & dosage , Cisplatin/pharmacology , Cyclooxygenase Inhibitors , Humans , Mass Spectrometry , Metabolomics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/pharmacology , Oxaliplatin , Thromboxane-A Synthase/antagonists & inhibitors , Tumor Cells, Cultured
19.
Curr Clin Pharmacol ; 5(3): 178-85, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20406172

ABSTRACT

Vascular disrupting agents (VDAs) represent a novel class of drugs targeting the tumor's blood supply. Conceptually and operationally different from currently used antiangiogenic agents, VDAs have a high specificity for the established but abnormal tumor vasculature. Upon administration, rapid changes in the microtubule cytoskeleton of tumor endothelial cells are induced, resulting in a cascade of events ultimately leading to blood flow stasis and vascular collapse. Subsequently, the cells in the core of the tumor become necrotic and die. However, tumor repopulation occurs from a rim of viable tumor tissue on the edges of the tumor, stimulating the search for appropriate combination strategies designed to interfere with the regrowth from the viable rim. Such combinations include chemotherapy, radiation, and antiangiogenic drugs. In recent years, understanding of the molecular and cellular mechanisms taking place in response to VDA therapy has improved substantially. Multiple drug combinations have been designed and tested in preclinical models, some of which have shown encouraging results. Clinical benefits are currently under investigation in a number of ongoing clinical trials, including randomized phase III trials.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neoplasms/blood supply , Neoplasms/drug therapy , Neovascularization, Pathologic , Angiogenesis Inhibitors/pharmacology , Combined Modality Therapy , Drug Delivery Systems , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...