Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2023 Feb 07.
Article in English | MEDLINE | ID: mdl-36798172

ABSTRACT

Most kidney cancers display evidence of metabolic dysfunction1-4 but how this relates to cancer progression in humans is unknown. We used a multidisciplinary approach to infuse 13C-labeled nutrients during surgical tumour resection in over 70 patients with kidney cancer. Labeling from [U-13C]glucose varies across cancer subtypes, indicating that the kidney environment alone cannot account for all metabolic reprogramming in these tumours. Compared to the adjacent kidney, clear cell renal cell carcinomas (ccRCC) display suppressed labelling of tricarboxylic acid (TCA) cycle intermediates in vivo and in organotypic slices cultured ex vivo, indicating that suppressed labeling is tissue intrinsic. Infusions of [1,2-13C]acetate and [U-13C]glutamine in patients, coupled with respiratory flux of mitochondria isolated from kidney and tumour tissue, reveal primary defects in mitochondrial function in human ccRCC. However, ccRCC metastases unexpectedly have enhanced labeling of TCA cycle intermediates compared to primary ccRCCs, indicating a divergent metabolic program during ccRCC metastasis in patients. In mice, stimulating respiration in ccRCC cells is sufficient to promote metastatic colonization. Altogether, these findings indicate that metabolic properties evolve during human kidney cancer progression, and suggest that mitochondrial respiration may be limiting for ccRCC metastasis but not for ccRCC growth at the site of origin.

3.
Front Oncol ; 11: 724682, 2021.
Article in English | MEDLINE | ID: mdl-34692501

ABSTRACT

INTRODUCTION: Patients with advanced germ cell tumors (GCT) receiving cisplatin-based chemotherapy have high rates of thromboembolic events (TEE) which can negatively affect their overall survival. While primary TEE prophylaxis during chemotherapy may prevent these events, it is unclear which patients will benefit in this setting. MATERIALS AND METHODS: A review of PubMed/Medline was conducted in December 2020 and all pertinent articles were evaluated for relevancy and quality of data for inclusion in the review. RESULTS: Studies on patients receiving initial cisplatin-based chemotherapy for advanced GCT have reported up to a 19% rate of TEE. This high rate may be associated with multiple factors including retroperitoneal lymphadenopathy, advanced clinical stage, high risk Khorana scores and presence of a central line. Large phase III clinical trials have demonstrated the benefit of low-molecular-weight-heparin and direct oral anticoagulants for primary prophylaxis and against recurrent TEE. However, primary prophylaxis is currently underutilized with GCT patients starting chemotherapy. CONCLUSION: Precise models to predict TEE risk and consideration of anticoagulation are difficult to develop owing to the relatively uncommon nature of GCT and lack of representation in primary TEE prophylaxis clinical trials. Despite these limitations, we believe that the benefits of prophylactic anticoagulation outweigh the risk of major bleeding in select GCT patients with higher risk of TEE. We have developed a simple algorithm to help guide TEE prophylaxis selection based on patient factors and route of chemotherapy administration. Given the high rate of TEE in GCT patients, we believe better utilization of primary prophylaxis in patient starting cisplatin-based chemotherapy will have clinical benefit.

4.
J Endocr Soc ; 5(1): bvaa158, 2021 Jan 01.
Article in English | MEDLINE | ID: mdl-33294761

ABSTRACT

Androgen deprivation therapy (ADT) is recommended for the treatment of advanced prostate cancer. Inadequate suppression of testosterone while on ADT poses a clinical challenge and requires evaluation of multiple potential causes, including adrenal virilizing disorders. We present 2 cases of elderly patients with prostate cancer who had undiagnosed congenital adrenal hyperplasia (CAH) driving persistent testosterone elevation during ADT. The first patient is a 73-year-old man who underwent radical prostatectomy on initial diagnosis and was later started on ADT with leuprolide following tumor recurrence. He had a testosterone level of 294.4 ng/dL and prostate-specific antigen (PSA) level of 17.7 ng/mL despite leuprolide use. Additional workup revealed adrenal nodular hyperplasia, elevated 17-hydroxyprogesterone (19 910 ng/dL) and dehydroepiandrosterone sulfate (378 mcg/dL), and 2 mutations of the CYP21A2 gene consistent with simple virilizing CAH. The second patient is an 82-year-old man who received stereotactic radiation therapy at time of diagnosis. He had insufficient suppression of testosterone with evidence of metastatic disease despite treatment with leuprolide and subsequently degarelix. Laboratory workup revealed elevated 17-hydroxyprogesterone (4910 ng/dL) and dehydroepiandrosterone sulfate (312 mcg/dL). Based on clinical, radiographic and biochemical findings, the patient was diagnosed with nonclassic CAH. The first patient initiated glucocorticoid therapy, and the second patient was treated with the CYP17 inhibitor abiraterone in combination with glucocorticoids. Both patients experienced rapid decline in testosterone and PSA levels. Inadequate testosterone suppression during ADT should trigger evaluation for causes of persistent hyperandrogenemia. CAH can lead to hyperandrogenemia and pose challenges when treating patients with prostate cancer.

5.
Clin Cancer Res ; 26(4): 793-803, 2020 02 15.
Article in English | MEDLINE | ID: mdl-31727677

ABSTRACT

PURPOSE: The heterodimeric transcription factor HIF-2 is arguably the most important driver of clear cell renal cell carcinoma (ccRCC). Although considered undruggable, structural analyses at the University of Texas Southwestern Medical Center (UTSW, Dallas, TX) identified a vulnerability in the α subunit, which heterodimerizes with HIF1ß, ultimately leading to the development of PT2385, a first-in-class inhibitor. PT2385 was safe and active in a first-in-human phase I clinical trial of patients with extensively pretreated ccRCC at UTSW and elsewhere. There were no dose-limiting toxicities, and disease control ≥4 months was achieved in 42% of patients. PATIENTS AND METHODS: We conducted a prospective companion substudy involving a subset of patients enrolled in the phase I clinical trial at UTSW (n = 10), who were treated at the phase II dose or above, involving multiparametric MRI, blood draws, and serial biopsies for biochemical, whole exome, and RNA-sequencing studies. RESULTS: PT2385 inhibited HIF-2 in nontumor tissues, as determined by a reduction in erythropoietin levels (a pharmacodynamic marker), in all but one patient, who had the lowest drug concentrations. PT2385 dissociated HIF-2 complexes in ccRCC metastases, and inhibited HIF-2 target gene expression. In contrast, HIF-1 complexes were unaffected. Prolonged PT2385 treatment resulted in the acquisition of resistance, and we identified a gatekeeper mutation (G323E) in HIF2α, which interferes with drug binding and precluded HIF-2 complex dissociation. In addition, we identified an acquired TP53 mutation elsewhere, suggesting a possible alternate mechanism of resistance. CONCLUSIONS: These findings demonstrate a core dependency on HIF-2 in metastatic ccRCC and establish PT2385 as a highly specific HIF-2 inhibitor in humans. New approaches will be required to target mutant HIF-2 beyond PT2385 or the closely related PT2977 (MK-6482).


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Carcinoma, Renal Cell/drug therapy , Indans/therapeutic use , Kidney Neoplasms/drug therapy , Sulfones/therapeutic use , Aged , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Clinical Trials, Phase I as Topic , Drug Resistance, Neoplasm , Female , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Male , Middle Aged , Multiparametric Magnetic Resonance Imaging/methods , Prospective Studies
7.
Nat Chem Biol ; 14(11): 1032-1042, 2018 11.
Article in English | MEDLINE | ID: mdl-30297875

ABSTRACT

α-Ketoglutarate (αKG) is a key node in many important metabolic pathways. The αKG analog N-oxalylglycine (NOG) and its cell-permeable prodrug dimethyloxalylglycine (DMOG) are extensively used to inhibit αKG-dependent dioxygenases. However, whether NOG interference with other αKG-dependent processes contributes to its mode of action remains poorly understood. Here we show that, in aqueous solutions, DMOG is rapidly hydrolyzed, yielding methyloxalylglycine (MOG). MOG elicits cytotoxicity in a manner that depends on its transport by monocarboxylate transporter 2 (MCT2) and is associated with decreased glutamine-derived tricarboxylic acid-cycle flux, suppressed mitochondrial respiration and decreased ATP production. MCT2-facilitated entry of MOG into cells leads to sufficiently high concentrations of NOG to inhibit multiple enzymes in glutamine metabolism, including glutamate dehydrogenase. These findings reveal that MCT2 dictates the mode of action of NOG by determining its intracellular concentration and have important implications for the use of (D)MOG in studying αKG-dependent signaling and metabolism.


Subject(s)
Amino Acids, Dicarboxylic/chemistry , Ketoglutaric Acids/chemistry , Monocarboxylic Acid Transporters/metabolism , Adenosine Triphosphate/chemistry , Animals , Biochemical Phenomena , Cattle , Cell Line, Tumor , Citric Acid Cycle , Gene Expression Profiling , Glutamine/metabolism , Humans , Hydrolysis , Inhibitory Concentration 50 , MCF-7 Cells , Metabolomics , Mice , Mitochondria/metabolism , Oxygen/chemistry , Puromycin/chemistry , Signal Transduction , Tricarboxylic Acids/chemistry
8.
Cell Metab ; 28(5): 793-800.e2, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30146487

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is the most common form of human kidney cancer. Histological and molecular analyses suggest that ccRCCs have significantly altered metabolism. Recent human studies of lung cancer and intracranial malignancies demonstrated an unexpected preservation of carbohydrate oxidation in the tricarboxylic acid (TCA) cycle. To test the capacity of ccRCC to oxidize substrates in the TCA cycle, we infused 13C-labeled fuels in ccRCC patients and compared labeling patterns in tumors and adjacent kidney. After infusion with [U-13C]glucose, ccRCCs displayed enhanced glycolytic intermediate labeling, suppressed pyruvate dehydrogenase flow, and reduced TCA cycle labeling, consistent with the Warburg effect. Comparing 13C labeling among ccRCC, brain, and lung tumors revealed striking differences. Primary ccRCC tumors demonstrated the highest enrichment in glycolytic intermediates and lowest enrichment in TCA cycle intermediates. Among human tumors analyzed by intraoperative 13C infusions, ccRCC is the first to demonstrate a convincing shift toward glycolytic metabolism.


Subject(s)
Carcinoma, Renal Cell/metabolism , Glucose/metabolism , Kidney Neoplasms/metabolism , Adult , Aged , Carbon Isotopes/metabolism , Carcinoma, Renal Cell/pathology , Citric Acid Cycle , Glycolysis , Humans , Kidney/metabolism , Kidney/pathology , Kidney Neoplasms/pathology , Middle Aged , Oxidation-Reduction
9.
J Clin Oncol ; 36(9): 867-874, 2018 03 20.
Article in English | MEDLINE | ID: mdl-29257710

ABSTRACT

Purpose The von Hippel-Lindau tumor suppressor is inactivated in the majority of clear cell renal cell carcinomas (ccRCCs), leading to inappropriate stabilization of hypoxia-inducible factor-2α (HIF-2α). PT2385 is a first-in-class HIF-2α antagonist. Objectives of this first-in-human study were to characterize the safety, pharmacokinetics, pharmacodynamics, and efficacy, and to identify the recommended phase II dose (RP2D) of PT2385. Patients and Methods Eligible patients had locally advanced or metastatic ccRCC that had progressed during one or more prior regimens that included a vascular endothelial growth factor inhibitor. PT2385 was administered orally at twice-per-day doses of 100 to 1,800 mg, according to a 3 + 3 dose-escalation design, followed by an expansion phase at the RP2D. Results The dose-escalation and expansion phases enrolled 26 and 25 patients, respectively. Patients were heavily pretreated, with a median of four (range, one to seven) prior therapies. No dose-limiting toxicity was observed at any dose. On the basis of safety, pharmacokinetic, and pharmacodynamic profiling, the RP2D was defined as 800 mg twice per day. PT2385 was well tolerated, with anemia (grade 1 to 2, 35%; grade 3, 10%), peripheral edema (grade 1 to 2, 37%; grade 3, 2%), and fatigue (grade 1 to 2, 37%; no grade 3 or 4) being the most common treatment-emergent adverse events. No patients discontinued treatment because of adverse events. Complete response, partial response, and stable disease as best response were achieved by 2%, 12%, and 52% of patients, respectively. At data cutoff, eight patients remained in the study, with 13 patients in the study for ≥ 1 year. Conclusion PT2385 has a favorable safety profile and is active in patients with heavily pretreated ccRCC, validating direct HIF-2α antagonism for the treatment of patients with ccRCC.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Carcinoma, Renal Cell/drug therapy , Indans/administration & dosage , Kidney Neoplasms/drug therapy , Sulfones/administration & dosage , Adult , Aged , Aged, 80 and over , Carcinoma, Renal Cell/blood , Carcinoma, Renal Cell/pathology , Dose-Response Relationship, Drug , Female , Humans , Indans/adverse effects , Indans/blood , Kidney Neoplasms/blood , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis , Sulfones/adverse effects , Sulfones/blood
10.
Clin Genitourin Cancer ; 13(2): 113-23, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25450031

ABSTRACT

BACKGROUND: The PTEN tumor suppressor is frequently lost in CRPC, with activation of Akt-mTOR signaling, driving growth. We conducted a phase I trial of the mTOR inhibitor, everolimus, and docetaxel in CRPC. PATIENTS AND METHODS: Eligible patients had progressive, metastatic, chemotherapy-naive CRPC. Patients received everolimus 10 mg daily for 2 weeks and underwent a restaging FDG-PET/computed tomography scan. Patient cohorts were subsequently treated at 3 dose levels of everolimus with docetaxel: 5 mg to 60 mg/m(2), 10 mg to 60 mg/m(2), and 10 mg to 70 mg/m(2). The primary end point was the safety and tolerability of combination therapy. RESULTS: Accrual was 4 patients at level 1, 3 patients at level 2, and 8 patients at level 3. Common toxicities were hematologic and fatigue. Serum concentrations of everolimus when administered with docetaxel were 1.5 to 14.8 ng/mL in patients receiving 5 mg everolimus and 4.5 to 55.4 ng/mL in patients receiving 10 mg everolimus. Four patients had partial metabolic response (PMR) using FDG-PET, 12 had stable metabolic disease, and 2 had progressive metabolic disease after a 2-week treatment with everolimus alone. Five of 12 evaluable patients experienced a prostate-specific antigen (PSA) reduction ≥ 50% during treatment with everolimus together with docetaxel. All 4 patients with a PMR according to PET imaging experienced a PSA reduction in response to everolimus with docetaxel, and 3 of 4 had PSA declines ≥ 50%. CONCLUSION: Everolimus 10 mg daily and docetaxel 60 mg/m(2) was safe in CRPC patients and these were the recommended doses in combination. FDG-PET response might serve as a biomarker for target inhibition by mTOR inhibitors.


Subject(s)
Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Everolimus/administration & dosage , Fluorodeoxyglucose F18/metabolism , Prostatic Neoplasms, Castration-Resistant/drug therapy , Taxoids/administration & dosage , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers, Tumor/metabolism , Docetaxel , Drug Administration Schedule , Everolimus/adverse effects , Everolimus/pharmacokinetics , Humans , Male , Maximum Tolerated Dose , Middle Aged , Positron-Emission Tomography/methods , Prostatic Neoplasms, Castration-Resistant/pathology , Taxoids/adverse effects , Taxoids/pharmacokinetics , Treatment Outcome
11.
BJU Int ; 110(11): 1729-35, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22928480

ABSTRACT

UNLABELLED: Study Type--Therapy (cohort) Level of Evidence 2a. What's known on the subject? and What does the study add? Despite expanding treatment options for castration-resistant prostate cancer (CRPC), therapies with long response duration remain intangible due to prostate cancer cells' natural ability to develop iterative resistance. Androgen receptor (AR) signaling has been shown to play a critical role in CRPC and its expression is regulated by the PI3K-Akt pathway. Thus inhibition of AR signalling and PI3K-Akt-mTOR (a downstream mediator of the PI3K-Akt pathway) pathway is a logical combination in CRPC and we report a phase II trial of RAD001 and bicalutamide. Our study is the first clinical trial report of an AR inhibitor of PI3K-Akt-mTOR. The AR pathway and the PI3K-Akt-mTOR pathway are two of the most relevant growth pathway for CRPC. Despite low efficacy results from our trial there will be significant interest in the field for these data (dose, schedule, response, toxicity, trial design) as newer generations of both AR inhibitors and PI3K-Akt-mTOR inhibitors are in development and likely will be tested in combination in CRPC. OBJECTIVES: • To determine best overall response and duration of response of RAD001, a selective inhibitor of mammalian target of rapamycin, in combination with bicalutamide in castration-resistant prostate cancer (CRPC). • To characterize the toxicity profile of RAD001 in combination with bicalutamide in patients with CRPC. PATIENTS AND METHODS: • A phase II study was conducted to explore the efficacy and tolerability of RAD001 (10 mg daily) in combination with bicalutamide (50 mg daily) in men with progressive CRPC. • The primary endpoint was a composite of prostate-specific antigen (PSA) level and measurable disease response by standard criteria. • This single-stage trial with a sample size of 38 eligible patients provided 90% power to differentiate a response rate of ≥ 40% from a response rate of ≤ 20%, as expected for bicalutamide alone (α= 0.10, power = 0.90). RESULTS: • In total, 36 men were enrolled, with a median (range) age of 68 (60-72) years and median (range) baseline PSA level of 22.2 (8.4-121.3) ng/mL, and 89% had metastatic disease. • There were 31 (86%) patients had previously used bicalutamide for a median duration of 7.4 months. • There were two patients with a confirmed PSA level decline ≥ 50%. • The median (interquartile range) time to progression was 8.7 (7.9-15.9) weeks. • The most common toxicity was grade 1/2 mucositis, which was observed in 20 (56%) patients. CONCLUSION: • The combination of RAD001 and bicalutamide in men with CRPC was well tolerated but had low activity and failed to achieve the primary endpoint of improved response compared to the results previously achieved for bicalutamide alone in this population.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Prostatic Neoplasms/drug therapy , Adenocarcinoma/blood , Aged , Androgen Receptor Antagonists/therapeutic use , Anilides/administration & dosage , Anilides/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Drug Resistance, Neoplasm , Everolimus , Humans , Male , Middle Aged , Nitriles/administration & dosage , Nitriles/adverse effects , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/blood , Sirolimus/administration & dosage , Sirolimus/adverse effects , Sirolimus/analogs & derivatives , Tosyl Compounds/administration & dosage , Tosyl Compounds/adverse effects , Treatment Outcome
12.
Nat Chem Biol ; 8(10): 839-47, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22922757

ABSTRACT

Cancer cells engage in a metabolic program to enhance biosynthesis and support cell proliferation. The regulatory properties of pyruvate kinase M2 (PKM2) influence altered glucose metabolism in cancer. The interaction of PKM2 with phosphotyrosine-containing proteins inhibits enzyme activity and increases the availability of glycolytic metabolites to support cell proliferation. This suggests that high pyruvate kinase activity may suppress tumor growth. We show that expression of PKM1, the pyruvate kinase isoform with high constitutive activity, or exposure to published small-molecule PKM2 activators inhibits the growth of xenograft tumors. Structural studies reveal that small-molecule activators bind PKM2 at the subunit interaction interface, a site that is distinct from that of the endogenous activator fructose-1,6-bisphosphate (FBP). However, unlike FBP, binding of activators to PKM2 promotes a constitutively active enzyme state that is resistant to inhibition by tyrosine-phosphorylated proteins. These data support the notion that small-molecule activation of PKM2 can interfere with anabolic metabolism.


Subject(s)
Biopolymers/metabolism , Cell Transformation, Neoplastic , Enzyme Activators/pharmacology , Pyruvate Kinase/metabolism , Animals , Biopolymers/chemistry , Blotting, Western , Cell Proliferation , Humans , Mice , Neoplasms/enzymology , Neoplasms/metabolism , Neoplasms/pathology , Pyruvate Kinase/chemistry
13.
Curr Opin Oncol ; 24(3): 272-7, 2012 May.
Article in English | MEDLINE | ID: mdl-22327837

ABSTRACT

PURPOSE OF REVIEW: The review examines recent advances in second-line hormonal therapy for the treatment of castrate-resistant prostate cancer (CRPC). RECENT FINDINGS: Recent data highlight the continued importance of androgen signaling in CRPC. These findings have spurred the development of novel inhibitors of adrenal and intra-tumoral androgen synthesis and novel androgen signaling inhibitors with activity in CRPC. In the past year abiraterone acetate, a CYP17 (17α-hydroxylase/17, 20 lyase) inhibitor, received US FDA approval for use in the treatment of metastatic CRPC in patients previously treated with docetaxel. Additionally, the novel androgen signaling inhibitor MDV3100 has been reported to confer a survival advantage compared to placebo in the same patient population. Here we review the scientific rationale for targeting androgen signaling in CRPC and the recent pivotal trials that support the use of novel second-line hormonal therapies. Additionally, we summarize ongoing preclinical and clinical efforts to ascertain and overcome mechanisms of resistance. SUMMARY: Novel inhibitors of extra-gonadal androgen synthesis and androgen receptor function demonstrate the continued importance of androgen signaling in CRPC. These agents have improved clinical outcomes for patients with metastatic CRPC.


Subject(s)
Androgen Antagonists/pharmacology , Androgens/metabolism , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm , Neoplasms, Hormone-Dependent/drug therapy , Prostatic Neoplasms/drug therapy , Androgen Antagonists/administration & dosage , Androgens/biosynthesis , Androstadienes/pharmacology , Androstenes , Androstenols/pharmacology , Antineoplastic Agents/administration & dosage , Benzamides , Benzimidazoles/pharmacology , Clinical Trials as Topic , Humans , Male , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Steroid 17-alpha-Hydroxylase/antagonists & inhibitors , Thiohydantoins/pharmacology
14.
J Clin Oncol ; 28(6): 1075-83, 2010 Feb 20.
Article in English | MEDLINE | ID: mdl-20085938

ABSTRACT

The phosphatidylinositol 3-kinase (PI3K) signaling axis impacts on cancer cell growth, survival, motility, and metabolism. This pathway is activated by several different mechanisms in cancers, including somatic mutation and amplification of genes encoding key components. In addition, PI3K signaling may serve integral functions for noncancerous cells in the tumor microenvironment. Consequently, therapeutics targeting the PI3K pathway are being developed at a rapid pace, and preclinical and early clinical studies are beginning to suggest specific strategies to effectively use them. However, the central role of PI3K signaling in a large array of diverse biologic processes raises concerns about its use in therapeutics and increases the need to develop sophisticated strategies for its use. In this review, we will discuss how PI3K signaling affects the growth and survival of tumor cells. From this vantage, we will consider how inhibitors of the PI3K signaling cascade, either alone or in combination with other therapeutics, can most effectively be used for the treatment of cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Animals , Humans , Phosphoinositide-3 Kinase Inhibitors
15.
Ther Adv Med Oncol ; 2(3): 209-19, 2010 May.
Article in English | MEDLINE | ID: mdl-21789135

ABSTRACT

Metastatic renal cell carcinoma (RCC) poses one of the great therapeutic challenges in oncology. RCC is predominantly refractory to treatment with traditional cytotoxic chemotherapies, and until recently management options were limited to immunotherapy or palliative care. However, in the past few years we have experienced a sea change in the treatment of advanced RCC with the introduction of targeted therapies that derive their efficacy at least in part through alterations in tumor angiogenesis. The tyrosine kinase inhibitors sunitinib, pazopanib, and sorafenib, the monoclonal antibody bevacizumab (in combination with interferon-α), and the rapamycin analogs, temsirolimus and everolimus, are now approved agents in the United States for the treatment of metastatic RCC. Efforts to expand upon these successes include developing novel antiangiogenic agents, optimizing concomitant and sequential regimens, identifying predictors of response to specific treatments, and further dissecting the underlying molecular pathogenesis of RCC to reveal novel therapeutic targets.

16.
Curr Oncol Rep ; 11(3): 218-26, 2009 May.
Article in English | MEDLINE | ID: mdl-19336014

ABSTRACT

The past few years have seen dramatic advances in the treatment of metastatic renal cell carcinoma (RCC). Dissection of the molecular pathways that regulate proliferation, apoptosis, and angiogenesis has led to the development of targeted therapies such as the receptor tyrosine kinase inhibitors sunitinib and sorafenib, the anti-vascular endothelial growth factor antibody bevacizumab, and a class of rapamycin analogues including everolimus and temsirolimus. Each of these agents has demonstrated clinical efficacy in patients with metastatic RCC. The challenge before us is to expand on these successes by identifying which patients will best respond to these targeted therapies, optimizing the proper combination or sequence of available therapies, developing agents with improved side effect profiles, and identifying novel therapeutic targets to expand our armamentarium in the treatment of RCC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Angiogenesis Inhibitors/therapeutic use , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/secondary , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Models, Biological , Receptors, Vascular Endothelial Growth Factor/metabolism , Signal Transduction/drug effects , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism
17.
Cell Metab ; 8(3): 179-81, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18762017

ABSTRACT

Phosphoinositide-3-OH kinases (PI3K) are critical regulators of cell metabolism, growth, and survival. In a recent publication in Nature, Jia et al. (2008) identify specific functions of the p110beta isoform of PI3K in glucose metabolism, cellular proliferation, and tumorigenesis.


Subject(s)
Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Protein Isoforms/metabolism , Animals , Cell Proliferation , Glucose/metabolism , Humans , Models, Biological , Neoplasms/genetics , Receptor Protein-Tyrosine Kinases/metabolism
18.
Perfusion ; 22(4): 299-301, 2007 Jul.
Article in English | MEDLINE | ID: mdl-18181520

ABSTRACT

A 36-year-old female with hemoglobin Nottingham (betaFG 5(98) Val --> Gly) causing severe hemolytic anemia and chronic thromboembolic pulmonary hypertension presented with symptomatic subacute right lower lobar pulmonary arterial thrombosis requiring surgical pulmonary thrombectomy. We describe a successful, multidisciplinary approach to the problems associated with this disease, particularly with the use of cardiopulmonary bypass and deep hypothermic circulatory arrest.


Subject(s)
Hemoglobinopathies/complications , Hemoglobins, Abnormal , Pulmonary Embolism/surgery , Thrombectomy , Adult , Cardiopulmonary Bypass , Circulatory Arrest, Deep Hypothermia Induced , Female , Humans , Hypertension, Pulmonary/complications , Pulmonary Embolism/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...