1
|
Kalot G, Godard A, Busser B, Bendellaa M, Dalonneau F, Paul C, Le Guével X, Josserand V, Coll JL, Denat F, Bodio E, Goze C, Gautier T, Sancey L. Lipoprotein interactions with water-soluble NIR-II emitting aza-BODIPYs boost the fluorescence signal and favor selective tumor targeting. Biomater Sci 2022; 10:6315-6325. [PMID: 36149672 DOI: 10.1039/d2bm01271e] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Following intravenous administration, the interaction of fluorescent exogenous molecules with circulating endogenous transporters can influence their photophysical properties as well as their fate and distribution, and possibly their recognition by different cell types. This type of interaction can be used to optimize the drug delivery but also the imaging properties of a compound of interest. In this study, we investigated the behavior of SWIR-WAZABY-01 fluorophore, a water-soluble aza-BODIPY dye emitting in the NIR-II region, both in vitro and in vivo. While the fluorescence emission of SWIR-WAZABY-01 was weak in aqueous solutions, it was intensely magnified in plasma (∼ ×30). Further analyses using lipoprotein gel electrophoresis and ultracentrifugation revealed interactions between SWIR-WAZABY-01 and plasma lipoproteins in vitro and ex vivo, in particular with LDL. The tumor uptake mechanism of SWIR-WAZABY-01 was investigated based on the presence of low-density lipoprotein (LDL) receptors and passive tumor uptake. Overall, we found that SWIR-WAZABY-01 interacts with lipoproteins enhancing their NIR-II fluorescence emission, and driving the tumor accumulation with regards to the expression of lipoprotein receptors (LDLR, SR-BI). Moreover, SWIR-WAZABY-01, by exploiting endogenous lipoproteins, arises as a new, potent and relevant tool to efficiently label LDL involved in pathologies.
Collapse
Affiliation(s)
- Ghadir Kalot
- Université Grenoble Alpes, Institute for Advanced Biosciences, INSERM U 1209, CNRS UMR 5309, 38000 Grenoble, France.
| | - Amélie Godard
- Institut de Chimie Moléculaire de l'Université de Bourgogne, Université Bourgogne Franche-Comté, CNRS UMR 6302, Dijon, France
| | - Benoit Busser
- Université Grenoble Alpes, Institute for Advanced Biosciences, INSERM U 1209, CNRS UMR 5309, 38000 Grenoble, France. .,Institut Universitaire de France (IUF), France.,Grenoble Alpes University Hospital (CHUGA), Grenoble, France
| | - Mohamed Bendellaa
- Université Grenoble Alpes, Institute for Advanced Biosciences, INSERM U 1209, CNRS UMR 5309, 38000 Grenoble, France.
| | - Fabien Dalonneau
- Université Grenoble Alpes, Institute for Advanced Biosciences, INSERM U 1209, CNRS UMR 5309, 38000 Grenoble, France.
| | - Catherine Paul
- Laboratoire d'Immunologie et Immunothérapie des Cancers, EPHE, PSL Research University, Université de Bourgogne, Dijon, France
| | - Xavier Le Guével
- Université Grenoble Alpes, Institute for Advanced Biosciences, INSERM U 1209, CNRS UMR 5309, 38000 Grenoble, France.
| | - Véronique Josserand
- Université Grenoble Alpes, Institute for Advanced Biosciences, INSERM U 1209, CNRS UMR 5309, 38000 Grenoble, France. .,OPTIMAL, Small animal Imaging Platform, 38000 Grenoble, France
| | - Jean-Luc Coll
- Université Grenoble Alpes, Institute for Advanced Biosciences, INSERM U 1209, CNRS UMR 5309, 38000 Grenoble, France.
| | - Franck Denat
- Institut de Chimie Moléculaire de l'Université de Bourgogne, Université Bourgogne Franche-Comté, CNRS UMR 6302, Dijon, France
| | - Ewen Bodio
- Institut de Chimie Moléculaire de l'Université de Bourgogne, Université Bourgogne Franche-Comté, CNRS UMR 6302, Dijon, France
| | - Christine Goze
- Institut de Chimie Moléculaire de l'Université de Bourgogne, Université Bourgogne Franche-Comté, CNRS UMR 6302, Dijon, France
| | - Thomas Gautier
- INSERM UMR1231, UFR Sciences de santé, Université Bourgogne Franche-Comté, Dijon, France
| | - Lucie Sancey
- Université Grenoble Alpes, Institute for Advanced Biosciences, INSERM U 1209, CNRS UMR 5309, 38000 Grenoble, France.
| |
Collapse
|
2
|
Charbe NB, Lagos CF, Ortiz CAV, Tambuwala M, Palakurthi SS, Zacconi FC. PCSK9 conjugated liposomes for targeted delivery of paclitaxel to the cancer cell: A proof-of-concept study. Biomed Pharmacother 2022; 153:113428. [PMID: 36076548 DOI: 10.1016/j.biopha.2022.113428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/27/2022] [Accepted: 07/14/2022] [Indexed: 11/02/2022] Open
|
3
|
Linking Late Endosomal Cholesterol with Cancer Progression and Anticancer Drug Resistance. Int J Mol Sci 2022; 23:ijms23137206. [PMID: 35806209 PMCID: PMC9267071 DOI: 10.3390/ijms23137206] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/22/2022] [Accepted: 06/25/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer cells undergo drastic metabolic adaptions to cover increased bioenergetic needs, contributing to resistance to therapies. This includes a higher demand for cholesterol, which often coincides with elevated cholesterol uptake from low-density lipoproteins (LDL) and overexpression of the LDL receptor in many cancers. This implies the need for cancer cells to accommodate an increased delivery of LDL along the endocytic pathway to late endosomes/lysosomes (LE/Lys), providing a rapid and effective distribution of LDL-derived cholesterol from LE/Lys to other organelles for cholesterol to foster cancer growth and spread. LDL-cholesterol exported from LE/Lys is facilitated by Niemann–Pick Type C1/2 (NPC1/2) proteins, members of the steroidogenic acute regulatory-related lipid transfer domain (StARD) and oxysterol-binding protein (OSBP) families. In addition, lysosomal membrane proteins, small Rab GTPases as well as scaffolding proteins, including annexin A6 (AnxA6), contribute to regulating cholesterol egress from LE/Lys. Here, we summarize current knowledge that links upregulated activity and expression of cholesterol transporters and related proteins in LE/Lys with cancer growth, progression and treatment outcomes. Several mechanisms on how cellular distribution of LDL-derived cholesterol from LE/Lys influences cancer cell behavior are reviewed, some of those providing opportunities for treatment strategies to reduce cancer progression and anticancer drug resistance.
Collapse
|
4
|
Vital CG, Maranhão RC, Freitas FR, Van Eyll BM, Graziani SR. Use of paclitaxel carried in lipid core nanoparticles in patients with late-stage solid cancers with bone metastases: Lack of toxicity and therapeutic benefits. J Bone Oncol 2022; 34:100431. [PMID: 35517058 PMCID: PMC9065304 DOI: 10.1016/j.jbo.2022.100431] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 11/30/2022] Open
Abstract
LDE-PTX at 175 mg/m2/3wk dose was devoid of toxicity at > 3rd line treatment. Among 18 LDE-PTX treated patients, 9 had PFS > 6 months and 2 had PFS > 1 yr. LDE-PTX diminished bone pain and improved quality of life in all treated patients.
Patients with heavily pretreated, late-stage cancer and bone metastasis are usually poor candidates for further chemotherapy. Previously, we showed that association to lipid nanoparticles (LDE) drastically decreases the toxicity of anti-cancer drugs. Here, we tested the hypothesis that paclitaxel (PTX) carried in LDE could benefit end-of-life patients with painful bone metastases that had been previously treated with conventional PTX. Methods: Eighteen consecutive patients with late-stage cancer, 8 with breast, 5 with prostate and 5 with lung carcinoma, aged 59±9 years, were included in this study. All were receiving opioid medication. LDE-PTX was administered at 175 mg/m 2 every 3 weeks until disease progression. Clinical imaging examinations and serum biochemistry determinations were performed to monitor disease progression. Intensity of bone pain, use of opioid medications and occurrence of pathological bone fractures were also evaluated. Results: In total, 104 chemotherapy cycles were performed and none of the patients showed clinical and laboratorial toxicities or pathological bone fractures. In all patients, pain was reduced so as to allow substitution of non-opioid for opioid medication. Median progression-free survival (PFS) was four months (95% CI 2.4-5.5), but in five patients PFS was longer than 6 months. Conclusions: Absence of observable clinical and laboratorial toxicities from LDE-PTX treatment, improvement of bone pain and the possible effect on PFS in some patients, despite previous use of conventional PTX, suggest that LDEPTX merits further clinical investigation.
Collapse
|
5
|
Jiang W, Hu JW, He XR, Jin WL, He XY. Statins: a repurposed drug to fight cancer. J Exp Clin Cancer Res 2021; 40:241. [PMID: 34303383 PMCID: PMC8306262 DOI: 10.1186/s13046-021-02041-2] [Citation(s) in RCA: 161] [Impact Index Per Article: 53.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022] Open
Abstract
As competitive HMG-CoA reductase (HMGCR) inhibitors, statins not only reduce cholesterol and improve cardiovascular risk, but also exhibit pleiotropic effects that are independent of their lipid-lowering effects. Among them, the anti-cancer properties of statins have attracted much attention and indicated the potential of statins as repurposed drugs for the treatment of cancer. A large number of clinical and epidemiological studies have described the anticancer properties of statins, but the evidence for anticancer effectiveness of statins is inconsistent. It may be that certain molecular subtypes of cancer are more vulnerable to statin therapy than others. Whether statins have clinical anticancer effects is still an active area of research. Statins appear to enhance the efficacy and address the shortcomings associated with conventional cancer treatments, suggesting that statins should be considered in the context of combined therapies for cancer. Here, we present a comprehensive review of the potential of statins in anti-cancer treatments. We discuss the current understanding of the mechanisms underlying the anti-cancer properties of statins and their effects on different malignancies. We also provide recommendations for the design of future well-designed clinical trials of the anti-cancer efficacy of statins.
Collapse
Affiliation(s)
- Wen Jiang
- Department of General Surgery, The Affiliated Provincial Hospital of Anhui Medical University, Hefei, 230001, P. R. China
| | - Jin-Wei Hu
- Department of General Surgery, The Affiliated Provincial Hospital of Anhui Medical University, Hefei, 230001, P. R. China
| | - Xu-Ran He
- Department of Finance, The First Affiliated Hospital of University of Science and Technology of China (Anhui Provincial Hospital), Hefei, 230001, P. R. China
| | - Wei-Lin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, P. R. China.
| | - Xin-Yang He
- Department of General Surgery, The First Affiliated Hospital of University of Science and Technology of China (Anhui Provincial Hospital), Hefei, 230001, P. R. China.
| |
Collapse
|
6
|
Mahboobnia K, Pirro M, Marini E, Grignani F, Bezsonov EE, Jamialahmadi T, Sahebkar A. PCSK9 and cancer: Rethinking the link. Biomed Pharmacother 2021; 140:111758. [PMID: 34058443 DOI: 10.1016/j.biopha.2021.111758] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/17/2021] [Accepted: 05/20/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Cancer is emerging as a major problem globally, as it accounts for the second cause of death despite medical advances. According to epidemiological and basic studies, cholesterol is involved in cancer progression and there are abnormalities in cholesterol metabolism of cancer cells including prostate, breast, and colorectal carcinomas. However, the importance of cholesterol in carcinogenesis and thereby the role of cholesterol homeostasis as a therapeutic target is still a debated area in cancer therapy. Proprotein convertase subtilisin/kexin type-9 (PCSK9), a serine protease, modulates cholesterol metabolism by attachment to the LDL receptor (LDLR) and reducing its recycling by targeting the receptor for lysosomal destruction. Published research has shown that PCSK9 is also involved in degradation of other LDLR family members namely very-low-density-lipoprotein receptor (VLDLR), lipoprotein receptor-related protein 1 (LRP-1), and apolipoprotein E receptor 2 (ApoER2). As a result, this protein represents an interesting therapeutic target for the treatment of hypercholesterolemia. Interestingly, clinical trials on PCSK9-specific monoclonal antibodies have reported promising results with high efficacy in lowering LDL-C and in turn reducing cardiovascular complications. It is important to note that PCSK9 mediates several other pathways apart from its role in lipid homeostasis, including antiviral activity, hepatic regeneration, neuronal apoptosis, and modulation of various signaling pathways. Furthermore, recent literature has illustrated that PCSK9 is closely associated with incidence and progression of several cancers. In a number of studies, PCSK9 siRNA was shown to effectively suppress the proliferation and invasion of the several studied tumor cells. Hence, a novel application of PCSK9 inhibitors/silencers in cancer/metastasis could be considered. However, due to poor data on effectiveness and safety of PCSK9 inhibitors in cancer, the impact of PCSK9 inhibition in these pathological conditions is still unknown. SEARCH METHODS A vast literature search was conducted to find intended studies from 1956 up to 2020, and inclusion criteria were original peer-reviewed publications. PURPOSE OF REVIEW To date, PCSK9 has been scantly investigated in cancer. The question that needs to be discussed is "How does PCSK9 act in cancer pathophysiology and what are the risks or benefits associated to its inhibition?". We reviewed the available publications highlighting the contribution of this proprotein convertase in pathways related to cancer, with focus on the potential implications of its long-term pharmacological inhibition in cancer therapy.
Collapse
Affiliation(s)
- Khadijeh Mahboobnia
- Department of Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Matteo Pirro
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Ettore Marini
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Francesco Grignani
- Unit of Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Evgeny E Bezsonov
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Institute of Human Morphology, 3 Tsyurupa Street, Moscow 117418, Russia; Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 8 Baltiiskaya Street, Moscow 125315, Russia
| | - Tannaz Jamialahmadi
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran; Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Feldt M, Menard J, Rosendahl AH, Lettiero B, Bendahl PO, Belting M, Borgquist S. The effect of statin treatment on intratumoral cholesterol levels and LDL receptor expression: a window-of-opportunity breast cancer trial. Cancer Metab 2020; 8:25. [PMID: 33292612 PMCID: PMC7682108 DOI: 10.1186/s40170-020-00231-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 10/27/2020] [Indexed: 12/23/2022] Open
Abstract
Background Deregulated lipid metabolism is common in cancer cells and the mevalonate pathway, which synthesizes cholesterol, is central in lipid metabolism. This study aimed to assess statin-induced changes of the intratumoral levels of cholesterol and the expression of the low-density lipoprotein receptor (LDLR) to enhance our understanding of the role of the mevalonate pathway in cancer cholesterol metabolism. Methods This study is based on a phase II clinical trial designed as a window-of-opportunity trial including 50 breast cancer patients treated with 80 mg of atorvastatin/day for 2 weeks, between the time of diagnosis and breast surgery. Lipids were extracted from frozen tumor tissue sampled pre- and post-atorvastatin treatment. Intratumoral cholesterol levels were measured using a fluorometric quantitation assay. LDLR expression was evaluated by immunohistochemistry on formalin-fixed paraffin-embedded tumor tissue. Paired blood samples pre- and post-atorvastatin were analyzed for circulating low-density lipoprotein (LDL), high-density lipoprotein (HDL), apolipoprotein A1, and apolipoprotein B. In vitro experiments on MCF-7 breast cancer cells treated with atorvastatin were performed for comparison on the cellular level. Results In the trial, 42 patients completed all study parts. From the paired tumor tissue samples, assessment of the cholesterol levels was achievable for 14 tumors, and for the LDLR expression in 24 tumors. Following atorvastatin treatment, the expression of LDLR was significantly increased (P = 0.004), while the intratumoral levels of total cholesterol remained stable. A positive association between intratumoral cholesterol levels and tumor proliferation measured by Ki-67 expression was found. In agreement with the clinical findings, results from in vitro experiments showed no significant changes of the intracellular cholesterol levels after atorvastatin treatment while increased expression of the LDLR was found, although not reaching statistical significance. Conclusions This study shows an upregulation of LDLR and preserved intratumoral cholesterol levels in breast cancer patients treated with statins. Together with previous findings on the anti-proliferative effect of statins in breast cancer, the present data suggest a potential role for LDLR in the statin-induced regulation of breast cancer cell proliferation. Trial registration The study has been registered at ClinicalTrials.gov (i.e., ID number: NCT00816244, NIH), December 30, 2008. Supplementary Information The online version contains supplementary material available at 10.1186/s40170-020-00231-8.
Collapse
Affiliation(s)
- Maria Feldt
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden. .,Department of Oncology, Skåne University Hospital, Lund, Sweden.
| | - Julien Menard
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden
| | - Ann H Rosendahl
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden.,Department of Oncology, Skåne University Hospital, Lund, Sweden
| | - Barbara Lettiero
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden
| | - Pär-Ola Bendahl
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden
| | - Mattias Belting
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden.,Department of Oncology, Skåne University Hospital, Lund, Sweden.,Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Signe Borgquist
- Department of Clinical Sciences Lund, Division of Oncology and Pathology, Lund University, Lund, Sweden.,Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
8
|
Role of cholesterol metabolism in the anticancer pharmacology of selective estrogen receptor modulators. Semin Cancer Biol 2020; 73:101-115. [PMID: 32931953 DOI: 10.1016/j.semcancer.2020.08.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/13/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022]
Abstract
Selective estrogen receptor modulators (SERMs) are a class of compounds that bind to estrogen receptors (ERs) and possess estrogen agonist or antagonist actions in different tissues. As such, they are widely used drugs. For instance, tamoxifen, the most prescribed SERM, is used to treat ERα-positive breast cancer. Aside from their therapeutic targets, SERMs have the capacity to broadly affect cellular cholesterol metabolism and handling, mainly through ER-independent mechanisms. Cholesterol metabolism reprogramming is crucial to meet the needs of cancer cells, and different key processes involved in cholesterol homeostasis have been associated with cancer progression. Therefore, the effects of SERMs on cholesterol homeostasis may be relevant to carcinogenesis, either by contributing to the anticancer efficacy of these compounds or, conversely, by promoting resistance to treatment. Understanding these aspects of SERMs actions could help to design more efficacious therapies. Herein we review the effects of SERMs on cellular cholesterol metabolism and handling and discuss their potential in anticancer pharmacology.
Collapse
|
9
|
Pan H, Sun Y, Cao D, Wang L. Low-density lipoprotein decorated and indocyanine green loaded silica nanoparticles for tumor-targeted photothermal therapy of breast cancer. Pharm Dev Technol 2019; 25:308-315. [PMID: 31820663 DOI: 10.1080/10837450.2019.1684944] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Hongying Pan
- Department of Thyroid and Breast Surgery, Danyang People’s Hospital, Danyang, Jiangsu, China
| | - Yi Sun
- Department of Thyroid and Breast Surgery, Danyang People’s Hospital, Danyang, Jiangsu, China
| | - Danxia Cao
- Department of Thyroid and Breast Surgery, Danyang People’s Hospital, Danyang, Jiangsu, China
| | - Lihui Wang
- Central Laboratory, Danyang People’s Hospital, Danyang, Jiangsu, China
| |
Collapse
|
10
|
Correlations between the metabolic profile and 18F-FDG-Positron Emission Tomography-Computed Tomography parameters reveal the complexity of the metabolic reprogramming within lung cancer patients. Sci Rep 2019; 9:16212. [PMID: 31700108 PMCID: PMC6838313 DOI: 10.1038/s41598-019-52667-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 10/12/2019] [Indexed: 12/15/2022] Open
Abstract
Several studies have demonstrated that the metabolite composition of plasma may indicate the presence of lung cancer. The metabolism of cancer is characterized by an enhanced glucose uptake and glycolysis which is exploited by 18F-FDG positron emission tomography (PET) in the work-up and management of cancer. This study aims to explore relationships between 1H-NMR spectroscopy derived plasma metabolite concentrations and the uptake of labeled glucose (18F-FDG) in lung cancer tissue. PET parameters of interest are standard maximal uptake values (SUVmax), total body metabolic active tumor volumes (MATVWTB) and total body total lesion glycolysis (TLGWTB) values. Patients with high values of these parameters have higher plasma concentrations of N-acetylated glycoproteins which suggest an upregulation of the hexosamines biosynthesis. High MATVWTB and TLGWTB values are associated with higher concentrations of glucose, glycerol, N-acetylated glycoproteins, threonine, aspartate and valine and lower levels of sphingomyelins and phosphatidylcholines appearing at the surface of lipoproteins. These higher concentrations of glucose and non-carbohydrate glucose precursors such as amino acids and glycerol suggests involvement of the gluconeogenesis pathway. The lower plasma concentration of those phospholipids points to a higher need for membrane synthesis. Our results indicate that the metabolic reprogramming in cancer is more complex than the initially described Warburg effect.
Collapse
|
11
|
Ao M, Xiao X, Ao Y. Low density lipoprotein modified silica nanoparticles loaded with docetaxel and thalidomide for effective chemotherapy of liver cancer. ACTA ACUST UNITED AC 2018. [PMID: 29513882 PMCID: PMC5912100 DOI: 10.1590/1414-431x20176650] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the present study, we successfully developed a docetaxel (DTX) and thalidomide (TDD) co-delivery system based on low density lipoprotein (LDL) modified silica nanoparticles (LDL/SLN/DTX/TDD). By employing the tumor homing property of LDL and the drug-loading capability of silica nanoparticles, the prepared LDL/SLN/DTX/TDD was expected to locate and specifically deliver the loaded drugs (DTX and TDD) to achieve effective chemotherapy of liver cancer. In vitro analysis revealed that nano-sized LDL/SLN/DTX/TDD with decent drug loading capabilities was able to increase the delivery efficiency by targeting the low density lipoprotein receptors, which were overexpressed on HepG2 human hepatocellular liver carcinoma cell line, which exerted better cytotoxicity than unmodified silica nanoparticles and free drugs. In vivo imaging and anti-cancer assays also confirmed the preferable tumor-homing and synergetic anti-cancer effects of LDL/SLN/DTX/TDD.
Collapse
|
12
|
Elevated tumor LDLR expression accelerates LDL cholesterol-mediated breast cancer growth in mouse models of hyperlipidemia. Oncogene 2017; 36:6462-6471. [PMID: 28759039 PMCID: PMC5690879 DOI: 10.1038/onc.2017.247] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/14/2017] [Accepted: 06/16/2017] [Indexed: 12/11/2022]
Abstract
Obesity is associated with an increase in cancer-specific mortality in women with breast cancer. Elevated cholesterol, particularly low-density lipoprotein cholesterol (LDL-C) is frequently seen in obese women. Here, we aimed to determine the importance of elevated circulating LDL, and LDL receptor (LDLR) expression in tumor cells, on the growth of breast cancer using mouse models of hyperlipidemia. We describe two novel immunodeficient mouse models of hyperlipidemia (Rag1−/−/LDLR−/− and Rag1−/−/ApoE (apolipoprotein E)−/− mice), in addition to established immunocompetent LDLR−/− and ApoE−/− mice. The mice were used to study the effects of elevated LDL-C in human triple negative (MDA-MB-231) and mouse Her2/Neu overexpressing (MCNeuA) breast cancers. Tumors derived from MCNeuA and MDA-MB-231 cells had high LDLR expression and formed larger tumors in mice with high circulating LDL-C concentrations than in mice with lower LDL-C. Silencing the LDLR in the tumor cells led to decreased growth of Her2Neu overexpressing tumors in LDLR−/− and ApoE−/− mice, with increased Caspase 3 cleavage. Additionally, in vitro, silencing the LDLR led to decreased cell survival in serum-starved conditions, associated with Caspase 3 cleavage. Examining publically available human datasets, we found that high LDLR expression in human breast cancers was associated with decreased recurrence-free survival, particularly in patients treated with systemic therapies. Overall, our results highlight the importance of the LDLR in the growth of triple negative and HER2 overexpressing breast cancers in the setting of elevated circulating LDL-C, which may be important contributing factors to the increased recurrence and mortality in obese women with breast cancer.
Collapse
|
13
|
Sobot D, Mura S, Rouquette M, Vukosavljevic B, Cayre F, Buchy E, Pieters G, Garcia-Argote S, Windbergs M, Desmaële D, Couvreur P. Circulating Lipoproteins: A Trojan Horse Guiding Squalenoylated Drugs to LDL-Accumulating Cancer Cells. Mol Ther 2017; 25:1596-1605. [PMID: 28606375 PMCID: PMC5498828 DOI: 10.1016/j.ymthe.2017.05.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 05/23/2017] [Accepted: 05/23/2017] [Indexed: 11/30/2022] Open
Abstract
Selective delivery of anticancer drugs to rapidly growing cancer cells can be achieved by taking advantage of their high receptor-mediated uptake of low-density lipoproteins (LDLs). Indeed, we have recently discovered that nanoparticles made of the squalene derivative of the anticancer agent gemcitabine (SQGem) strongly interacted with the LDLs in the human blood. In the present study, we showed both in vitro and in vivo that such interaction led to the preferential accumulation of SQGem in cancer cells (MDA-MB-231) with high LDL receptor expression. As a result, an improved pharmacological activity has been observed in MDA-MB-231 tumor-bearing mice, an experimental model with a low sensitivity to gemcitabine. Accordingly, we proved that the use of squalene moieties not only induced the gemcitabine insertion into lipoproteins, but that it could also be exploited to indirectly target cancer cells in vivo.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/pathology
- Adenocarcinoma/therapy
- Animals
- Antineoplastic Agents/chemistry
- Antineoplastic Agents/pharmacology
- Breast Neoplasms/genetics
- Breast Neoplasms/pathology
- Breast Neoplasms/therapy
- Cell Line, Tumor
- Deoxycytidine/analogs & derivatives
- Deoxycytidine/chemistry
- Deoxycytidine/pharmacology
- Drug Carriers
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Lipoproteins, LDL/chemistry
- Lipoproteins, LDL/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Nude
- Mice, SCID
- Nanoparticles/administration & dosage
- Nanoparticles/chemistry
- Receptors, LDL/genetics
- Receptors, LDL/metabolism
- Squalene/chemistry
- Tumor Burden/drug effects
- Xenograft Model Antitumor Assays
- Gemcitabine
Collapse
Affiliation(s)
- Dunja Sobot
- Institut Galien Paris-Sud, UMR 8612, CNRS, University Paris-Sud, Université Paris-Saclay, Faculté de Pharmacie, 5 rue Jean-Baptiste Clément, 92296 Châtenay-Malabry Cedex, France
| | - Simona Mura
- Institut Galien Paris-Sud, UMR 8612, CNRS, University Paris-Sud, Université Paris-Saclay, Faculté de Pharmacie, 5 rue Jean-Baptiste Clément, 92296 Châtenay-Malabry Cedex, France
| | - Marie Rouquette
- Institut Galien Paris-Sud, UMR 8612, CNRS, University Paris-Sud, Université Paris-Saclay, Faculté de Pharmacie, 5 rue Jean-Baptiste Clément, 92296 Châtenay-Malabry Cedex, France
| | - Branko Vukosavljevic
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, Campus E8 1, 66123 Saarbruecken, Germany
| | - Fanny Cayre
- Institut Galien Paris-Sud, UMR 8612, CNRS, University Paris-Sud, Université Paris-Saclay, Faculté de Pharmacie, 5 rue Jean-Baptiste Clément, 92296 Châtenay-Malabry Cedex, France
| | - Eric Buchy
- Institut Galien Paris-Sud, UMR 8612, CNRS, University Paris-Sud, Université Paris-Saclay, Faculté de Pharmacie, 5 rue Jean-Baptiste Clément, 92296 Châtenay-Malabry Cedex, France
| | - Grégory Pieters
- SCBM, CEA, Université Paris Saclay, LabEx LERMIT, 91191 Gif-sur-Yvette, France
| | | | - Maike Windbergs
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Center for Infection Research, Campus E8 1, 66123 Saarbruecken, Germany; Institute of Pharmaceutical Technology, Buchmann Institute for Molecular Life Sciences, Goethe University, Max-von-Laue-Strasse 15, 60438 Frankfurt am Main, Germany
| | - Didier Desmaële
- Institut Galien Paris-Sud, UMR 8612, CNRS, University Paris-Sud, Université Paris-Saclay, Faculté de Pharmacie, 5 rue Jean-Baptiste Clément, 92296 Châtenay-Malabry Cedex, France
| | - Patrick Couvreur
- Institut Galien Paris-Sud, UMR 8612, CNRS, University Paris-Sud, Université Paris-Saclay, Faculté de Pharmacie, 5 rue Jean-Baptiste Clément, 92296 Châtenay-Malabry Cedex, France.
| |
Collapse
|
14
|
|
15
|
Abstract
PURPOSE OF REVIEW Adipocytes have adapted to store energy in the form of lipid and also secrete circulating factors called adipokines that signal to other tissues to coordinate energy homeostasis. These functions are disrupted in the setting of obesity, promoting the development of diseases such as diabetes, cardiovascular disease, and cancer. RECENT FINDINGS Obesity is linked to an increased risk of many types of cancer and increased cancer-related mortality. The basis for the striking association between obesity and cancer is not well understood. Here, we review the cellular and molecular pathways that appear to be involved in obesity-driven cancer. We also describe possible therapeutic considerations and highlight important unanswered questions in the field.
Collapse
Affiliation(s)
- Sarah E Ackerman
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Olivia A Blackburn
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - François Marchildon
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA.
- The Rockefeller University, 1230 York Avenue, Box 223, New York, NY, 10065, USA.
| |
Collapse
|
16
|
Kretzer IF, Maria DA, Guido MC, Contente TC, Maranhão RC. Simvastatin increases the antineoplastic actions of paclitaxel carried in lipid nanoemulsions in melanoma-bearing mice. Int J Nanomedicine 2016; 11:885-904. [PMID: 27022257 PMCID: PMC4788363 DOI: 10.2147/ijn.s88546] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
PURPOSE Lipid nanoemulsions (LDEs) that bind to low-density lipoprotein (LDL) receptors used as carriers of paclitaxel (PTX) can decrease toxicity and increase PTX antitumoral action. The administration of simvastatin (Simva), which lowers LDL-cholesterol, was tested as an adjuvant to commercial PTX and to PTX associated with LDE (LDE-PTX). MATERIALS AND METHODS B16F10 melanoma-bearing mice were treated with saline solution or LDE (controls), Simva, PTX, PTX and Simva, LDE-PTX, and LDE-PTX and Simva: PTX dose 17.5 μmol/kg (three intraperitoneal injections, 3 alternate days): Simva 50 mg/kg/day by gavage, 9 consecutive days. RESULTS Compared with saline controls, 95% tumor-growth inhibition was achieved by LDE-PTX and Simva, 61% by LDE-PTX, 44% by PTX and Simva, and 43% by PTX. Simva alone had no effect. Metastasis developed in only 37% of the LDE-PTX and Simva, 60% in LDE-PTX, and 90% in PTX and Simva groups. Survival rates were higher in LDE-PTX and Simva and in LDE-PTX groups. The LDE-PTX and Simva group presented tumors with reduced cellular density and increased collagen fibers I and III. Tumors from all groups showed reduction in immunohistochemical expression of ICAM, MCP-1, and MMP-9; LDE-PTX and Simva presented the lowest MMP-9 expression. Expression of p21 was increased in the Simva, LDE-PTX, and LDE-PTX and Simva groups. In the Simva and LDE-PTX and Simva groups, expression of cyclin D1, a proliferation and survival promoter of tumor cells, was decreased. Therapy with LDE-PTX and Simva showed negligible toxicity compared with PTX and Simva, which resulted in weight loss and myelosuppression. CONCLUSION Simva increased the antitumor activity of PTX carried in LDE but not of PTX commercial presentation, possibly because statins increase the expression of LDL receptors that internalize LDE-PTX.
Collapse
Affiliation(s)
- Iara F Kretzer
- Laboratory of Metabolism and Lipids, Heart Institute of the Medical School Hospital, University of São Paulo, São Paulo, Brazil; Department of Clinical Chemistry, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Durvanei A Maria
- Biochemistry and Biophysics Laboratories, Butantan Institute, São Paulo, Brazil
| | - Maria C Guido
- Laboratory of Metabolism and Lipids, Heart Institute of the Medical School Hospital, University of São Paulo, São Paulo, Brazil
| | - Thaís C Contente
- Laboratory of Metabolism and Lipids, Heart Institute of the Medical School Hospital, University of São Paulo, São Paulo, Brazil
| | - Raul C Maranhão
- Laboratory of Metabolism and Lipids, Heart Institute of the Medical School Hospital, University of São Paulo, São Paulo, Brazil; Department of Clinical Chemistry, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
17
|
Ben-Shmuel S, Rostoker R, Scheinman EJ, LeRoith D. Metabolic Syndrome, Type 2 Diabetes, and Cancer: Epidemiology and Potential Mechanisms. Handb Exp Pharmacol 2016; 233:355-372. [PMID: 25903410 DOI: 10.1007/164_2015_12] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Obesity is associated with multiple metabolic disorders that drive cardiovascular disease, T2D and cancer. The doubling in the number of obese adults over the past 3 decades led to the recognition of obesity as a "disease". With over 42 million children obese or overweight, this epidemic is rapidly growing worldwide. Obesity and T2D are both associated together and independently with an increased risk for cancer and a worse prognosis. Accumulating evidence from epidemiological studies revealed potential factors that may explain the association between obesity-linked metabolic disorders and cancer risk. Studies based on the insulin resistance MKR mice, highlighted the roe of the insulin receptor and its downstream signaling proteins in mediating hyperinsulinemia's mitogenic effects. Hypercholesterolemia was also shown to promote the formation of larger tumors and enhancement in metastasis. Furthermore, the conversion of cholesterol into 27-Hydroxycholesterol was found to link high fat diet-induced hypercholesterolemia with cancer pathophysiology. Alteration in circulating adipokines and cytokines are commonly found in obesity and T2D. Adipokines are involved in tumor growth through multiple mechanisms including mTOR, VEGF and cyclins. In addition, adipose tissues are known to recruit and alter macrophage phenotype; these macrophages can promote cancer progression by secreting inflammatory cytokines such as TNF-α and IL-6. Better characterization on the above factors and their downstream effects is required in order to translate the current knowledge into the clinic, but more importantly is to understand which are the key factors that drive cancer in each patient. Until we reach this point, policies and activities toward healthy diets and physical activities remain the best medicine.
Collapse
Affiliation(s)
- Sarit Ben-Shmuel
- Clinical Research Institute at Rambam (CRIR), Diabetes and Metabolism Clinical Research Center of Excellence, Rambam Health Care Campus, Haifa, Israel
| | - Ran Rostoker
- Clinical Research Institute at Rambam (CRIR), Diabetes and Metabolism Clinical Research Center of Excellence, Rambam Health Care Campus, Haifa, Israel
| | - Eyal J Scheinman
- Clinical Research Institute at Rambam (CRIR), Diabetes and Metabolism Clinical Research Center of Excellence, Rambam Health Care Campus, Haifa, Israel
| | - Derek LeRoith
- Clinical Research Institute at Rambam (CRIR), Diabetes and Metabolism Clinical Research Center of Excellence, Rambam Health Care Campus, Haifa, Israel.
| |
Collapse
|
18
|
Gallagher EJ, LeRoith D. Obesity and Diabetes: The Increased Risk of Cancer and Cancer-Related Mortality. Physiol Rev 2015; 95:727-48. [PMID: 26084689 DOI: 10.1152/physrev.00030.2014] [Citation(s) in RCA: 476] [Impact Index Per Article: 52.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Obesity and type 2 diabetes are becoming increasingly prevalent worldwide, and both are associated with an increased incidence and mortality from many cancers. The metabolic abnormalities associated with type 2 diabetes develop many years before the onset of diabetes and, therefore, may be contributing to cancer risk before individuals are aware that they are at risk. Multiple factors potentially contribute to the progression of cancer in obesity and type 2 diabetes, including hyperinsulinemia and insulin-like growth factor I, hyperglycemia, dyslipidemia, adipokines and cytokines, and the gut microbiome. These metabolic changes may contribute directly or indirectly to cancer progression. Intentional weight loss may protect against cancer development, and therapies for diabetes may prove to be effective adjuvant agents in reducing cancer progression. In this review we discuss the current epidemiology, basic science, and clinical data that link obesity, diabetes, and cancer and how treating obesity and type 2 diabetes could also reduce cancer risk and improve outcomes.
Collapse
Affiliation(s)
| | - Derek LeRoith
- Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
19
|
Pires LA, Hegg R, Freitas FR, Tavares ER, Almeida CP, Baracat EC, Maranhão RC. Effect of neoadjuvant chemotherapy on low-density lipoprotein (LDL) receptor and LDL receptor-related protein 1 (LRP-1) receptor in locally advanced breast cancer. Braz J Med Biol Res 2012; 45:557-64. [PMID: 22570085 PMCID: PMC3854302 DOI: 10.1590/s0100-879x2012007500068] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2011] [Accepted: 03/26/2012] [Indexed: 11/22/2022] Open
Abstract
Low-density lipoprotein (LDL) receptors are overexpressed in most neoplastic cell lines and provide a mechanism for the internalization and concentration of drug-laden nanoemulsions that bind to these receptors. The aim of the present study was to determine whether the administration of standard chemotherapeutic schemes can alter the expression of LDL and LDL receptor-related protein 1 (LRP-1) receptors in breast carcinoma. Fragments of tumoral and normal breast tissue from 16 consecutive volunteer women with breast cancer in stage II or III were obtained from biopsies before the beginning of neoadjuvant chemotherapy and after chemotherapy, from fragments excised during mastectomy. Tissues were analyzed by immunohistochemistry for both receptors. Because complete response to treatment was achieved in 4 patients, only the tumors from 12 were analyzed. Before chemotherapy, there was overexpression of LDL receptor in the tumoral tissue compared to normal breast tissue in 8 of these patients. LRP-1 receptor overexpression was observed in tumors of 4 patients. After chemotherapy, expression of both receptors decreased in the tumors of 6 patients, increased in 4 and was unchanged in 2. Nonetheless, even when chemotherapy reduced receptors expression, the expression was still above normal. The fact that chemotherapy does not impair LDL receptors expression supports the use of drug carrier systems that target neoplastic cells by the LDL receptor endocytic pathway in patients on conventional chemotherapy.
Collapse
Affiliation(s)
- L A Pires
- Laboratório de Metabolismo de Lípides, Instituto do Coração, Faculdade de Medicina, Hospital das Clínicas, Universidade de São Paulo, Brasil
| | | | | | | | | | | | | |
Collapse
|
20
|
Valduga CJ, Fernandes DC, Lo Prete AC, Azevedo CHM, Rodrigues DG, Maranhão RC. Use of a cholesterol-rich microemulsion that binds to low-density lipoprotein receptors as vehicle for etoposide. J Pharm Pharmacol 2010; 55:1615-22. [PMID: 14738586 DOI: 10.1211/0022357022232] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Abstract
A cholesterol-rich microemulsion (LDE) that binds to low-density lipoprotein (LDL) receptors is selectively taken up by malignant cells that overexpress those receptors and may be used as vehicle for antineoplastic agents. This study aimed to develop the association of etoposide with LDE. It was firstly observed that etoposide poorly associates with the microemulsion, therefore the experiments were performed with a lipophilic fatty acid derivative of the drug. The association of etoposide oleate with LDE was almost 100% and was tested for physical and chemical stability, as well as for cellular uptake, toxicity in mice and cytotoxic activity against a neoplastic cell line (NCI-H292). Uptake and cytotoxic activity of LDE-etoposide oleate by NCI-H292 cells was mediated by LDL receptors. The anti-proliferative activity of LDE-etoposide oleate against the neoplastic cells was smaller than that of etoposide oleate (IC50 (drug concentration required to inhibit 50% of the cell growth) = 0.48 and 0.19 mm, respectively). This difference, however, can be ascribed to the activity of the commercially used vehicle and not the drug itself because when this vehicle was added to the cultures with LDE-etoposide oleate, the IC50 decreased. On the other hand, the tolerability of LDE-etoposide oleate to mice was remarkable, such that its lethal dose (LD50) was about five-fold that of the commercial formulation (LD50=315 and 58 mg kg−1, respectively). In conclusion, LDE-etoposide oleate association is stable and the cytostatic activity of the drug is preserved while its toxicity to animals is small. By diminishing the side effects and directing etoposide to neoplastic tissues, LDE may be regarded as an advance in chemotherapy with this drug.
Collapse
Affiliation(s)
- Claudete J Valduga
- Lipid Metabolism Laboratory, the Heart Institute (InCor) of the Medical School Hospital, São Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
21
|
Kim TG, Kang SY, Kang JH, Cho MY, Kim JI, Kim SH, Kim JS. Gene transfer into human hepatoma cells by receptor-associated protein/polylysine conjugates. Bioconjug Chem 2004; 15:326-32. [PMID: 15025528 DOI: 10.1021/bc0340262] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Receptor-associated protein (RAP) is a ligand for all members of low-density lipoprotein (LDL) receptor families. RAP is internalized into cells via receptor-mediated endocytic trafficking, making it an attractive mechanism for efficient gene delivery. In this study, we have developed a gene delivery system using RAP as a targeting ligand. A RAP cDNA lacking a C-terminal heparin-binding domain was amplified by polymerase chain reaction (PCR) from a human liver cDNA library and was reamplified by using a primer containing a cysteine codon at its carboxyl end to facilitate its conjugation to polylysine (polyK). RAP was purified using a bacterial expression system and coupled to poly-D-lysine (PDL) or poly-L-lysine (PLL) of average MW 50 kDa via the heterobifunctional cross-linker SPDP. Using fluorescence-labeled RAP ligand, cellular uptake of the transfection complexes into HepG2 cells was shown to be highly efficient and more specific to PDL-conjugated RAP compared with PLL-conjugated one. Plasmid DNA containing a luciferase reporter gene was condensed with either RAP-PDL or RAP-PLL. In vitro transfection into HepG2 cells with RAP-PDL conjugate resulted in significantly higher luciferase expression levels in comparison to either nonconjugated PDL, or RAP-PLL, or LipofecAMINE/DNA complexes in the presence of 10% fetal bovine serum. Luciferase expression was inhibited by the addition of excess RAP. Treatment of the cells with Lovastatin, which inhibits HMG-Co reductase and increases expression of LDL receptor, stimulates luciferase expression, suggesting that the gene delivery is specifically mediated by LDL receptor. Thus, RAP-PDL conjugates have the potential to be used as a new nonviral gene delivery vector.
Collapse
Affiliation(s)
- Tae-Gyun Kim
- Department of Pharmacology, National Institute of Toxicological Research, Korea Food and Drug Administration, Seoul 122-704, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
22
|
Gonçalves RP, Hungria VTM, Chiattone CS, Pozzi DB, Maranhão RC. Metabolism of chylomicron-like emulsions in patients with Hodgkin's and with non-Hodgkin's lymphoma. Leuk Res 2003; 27:147-53. [PMID: 12526920 DOI: 10.1016/s0145-2126(02)00087-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND Chylomicrons carry in the bloodstream dietary fats absorbed the intestine for storage in the body tissues such as adipose and muscle. The two-step chylomicron metabolism consists in lipolysis by lipoprotein lipase on vessel walls and hepatic uptake of triglyceride-depleted remnants. Chylomicron metabolism is understudied in cancer, despite its direct involvement in the patient nutritional status. We investigated this metabolism in Hodgkin and non-Hodgkin lymphoma patients, using the method of triglyceride-rich emulsions that mimic chylomicrons. PATIENTS AND METHODS The chylomicron-like emulsion, labeled with [9,10-3H]glycerol-trioleate and [1-14C] cholesteryl-oleate was intravenously injected into 11 Hodgkin's, 19 non-Hodgkin's patients and 12 healthy subjects. Triglyceride kinetics evaluate lipolysis whereas cholesteryl ester kinetics evaluate remnant removal. RESULTS Plasma total, LDL, HDL cholesterol, apo B, apo A1 and Lp(a) values were similar between the three groups, but VLDL cholesterol and triglycerides were higher in the lymphoma groups. The fractional catabolic rate (FCR, in min(-1)) of the emulsion triglycerides was roughly three-fold smaller in non-Hodgkin's (0.043+/-0.007, mean+/-S.E.M., P<0.001) and Hodgkin's (0.045+/-0.009, P<0.0001) lymphoma patients compared with the control values (0.151+/-0.032). FCR of the emulsion cholesteryl esters, was four-fold smaller in non-Hodgkin's (0.016+/-0.002, P<0.0001), and three-fold in Hodgkin's lymphoma patients (0.024+/-0.006, P<0.001) compared with the control group (0.069+/-0.013). The lipolysis index, calculated from the decay curves of both isotopes was also markedly smaller in both groups of lymphoma patients compared with the controls. CONCLUSIONS In both lymphoma groups, marked alterations in chylomicron lipolysis and remnant removal occurs.
Collapse
Affiliation(s)
- Romélia P Gonçalves
- Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | | | | | | | | |
Collapse
|
23
|
Rodrigues DG, Covolan CC, Coradi ST, Barboza R, Maranhão RC. Use of a cholesterol-rich emulsion that binds to low-density lipoprotein receptors as a vehicle for paclitaxel. J Pharm Pharmacol 2002; 54:765-72. [PMID: 12078992 DOI: 10.1211/0022357021779104] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
A cholesterol-rich emulsion (LDE) is taken up by malignant cells which over-express low-density lipoprotein (LDL) receptors and thus may be used as a carrier for drugs directed against neoplastic cells. In this study, we associated the antineoplastic agent paclitaxel to LDE and analysed the new formulation's incorporation efficiency, chemical and physical stability, cellular uptake and cytostatic activity against a neoplastic cell line and the acute toxicity to rats. A paclitaxel incorporation efficiency of approximately 75% was achieved when paclitaxel was mixed with LDE at a 6:1 lipid-to-drug molar ratio. The association of paclitaxel with LDE increased by 54% the mean diameter of the emulsion particles but did not damage the paclitaxel chemical structure as analysed by HPLC. Results from gradient ultracentrifugation and Sephadex G25 gel filtration indicated that the binding of the drug to the emulsion was stable. It was shown that the cellular uptake and the cytotoxic activity of LDE-paclitaxel by a neoplastic cell line (NCI-H292 cells) was indeed mediated by the LDL receptors. The antiproliferative activity of LDE-paclitaxel against NCI-H292 cells was less than that of a commercial paclitaxel preparation (50% inhibitory concentration, IC50 = 2.60 and 0.45 microM, respectively). This difference, however, can be ascribed to the in-vitro anti-proliferative activity of the commercial paclitaxel vehicle Cremophor EL; when Cremophor EL was added to the cultures with LDE-paclitaxel, the IC50 value was reduced to 0.45 microM, attaining that of the commercial paclitaxel preparation. The tolerability of LDE-paclitaxel in rats was remarkable, such that its lethal dose (LD50) was ten-fold greater than that of the commercial formulation (LD50 = 324 and 31.8 mg kg(-1), respectively). Therefore, LDE-paclitaxel association is stable and the cytostatic activity of the drug is preserved while its toxicity to rats is small. By diminishing the side effects and directing paclitaxel to neoplastic tissues, LDE may be useful as adjuvant in chemotherapy with this drug.
Collapse
Affiliation(s)
- Debora G Rodrigues
- Lipid Metabolism Laboratory, the Heart Institute (INCOR) of the Medical School Hospital, University of São Paulo, Brazil
| | | | | | | | | |
Collapse
|
24
|
Ades A, Carvalho JP, Graziani SR, Amancio RF, Souen JS, Pinotti JA, Maranhão RC. Uptake of a cholesterol-rich emulsion by neoplastic ovarian tissues. Gynecol Oncol 2001; 82:84-7. [PMID: 11426966 DOI: 10.1006/gyno.2001.6203] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Previously, it was shown that a lipidic emulsion (LDE) composed of phospholipids and cholesterol esters which binds to low-density lipoprotein (LDL) receptors may concentrate in acute myeloid leukemia cells. In this study, we aimed to verify whether LDE also has the ability to concentrate in malignant ovarian cancer after being injected into the blood circulation of the patients. METHODS Three groups of women scheduled for surgery were included in the survey: 13 bearing malignant tumors, 9 with benign ovarian tumors, and 13 without ovarian tumor who were scheduled to undergo oophorectomy due to malignant disease of the uterine cervix or endometrium. On the day prior to surgery they were injected with LDE labeled with [(14)C]cholesteryl oleate. Specimens of tumors and normal ovaries excised during surgery were lipid extracted and analyzed for radioactivity counting. Results were expressed in radioactive count (cpm) per gram of tissue. RESULTS The mean of the uptakes of the emulsion radioactivity by the malignant tumors was roughly eightfold greater when compared with that of the contralateral normal ovaries (2261 +/- 1444 and 275 +/- 137 cpm/g, respectively, P < 0.012), benign tumors, and normal ovaries of the patients without ovarian tumors. CONCLUSION LDE has the ability to concentrate in malignant ovarian tumor tissue. Therefore, it can be used as a vehicle to direct cytotoxic drugs against malignant ovarian tumors, thus diminishing the side effects of chemotherapy.
Collapse
Affiliation(s)
- A Ades
- Department of Obstetrics and Gynecology, University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | |
Collapse
|
25
|
Mehta N, Hordines J, Volpe C, Doerr R, Cohen SA. Cellular effects of hypercholesterolemia in modulation of cancer growth and metastasis: a review of the evidence. Surg Oncol 1997; 6:179-85. [PMID: 9576633 DOI: 10.1016/s0960-7404(97)00027-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hypercholesterolemia and increased cancer risk have been associated, particularly with the high fat diets characteristic of Western societies. We were interested in the possible association between preexisting hypercholesterolemia and the rapidity and extent of tumor metastases in these patients. To date there has been only a few studies that have suggested and explored this determinant of cancer metastases although it may play a role in a subset of patients who develop cancers. This article will review the literature on the effects of LDL-cholesterol on cell proliferation and differentiation and speculate on mechanisms of involvement of a hypercholesterolemic milieu on cancer progression and enhancement of metastatic potential.
Collapse
Affiliation(s)
- N Mehta
- Department of Surgery, VA Medical Center, State University of New York at Buffalo, 14215, USA
| | | | | | | | | |
Collapse
|
26
|
Mannello F, Bocchiotti G, Pignatti Morano F, Fratepietro L, Gazzanelli G. Lipids status in human breast cyst fluids. Cancer Lett 1996. [DOI: 10.1016/s0304-3835(06)80023-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
27
|
Jasanada F, Urizzi P, Souchard JP, Le Gaillard F, Favre G, Nepveu F. Indium-111 labeling of low density lipoproteins with the DTPA-bis(stearylamide): evaluation as a potential radiopharmaceutical for tumor localization. Bioconjug Chem 1996; 7:72-81. [PMID: 8741993 DOI: 10.1021/bc950073l] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
In order to use the LDL receptor pathway to target radionuclides to cancer sites for imaging and diagnostic purposes, a labeling procedure of LDL with 111In using the DTPA-bis(stearylamide) (L) has been developed. This bifunctional ligand is intended to be incorporated into the phospholipid monolayer of LDL and to specifically chelate the In3+ cation at the surface. The ligand was incorporated into LDL in buffered medium with a 65-80% yield. The L-LDL samples are stable over a 24 h period when examined by dialysis, allowing their storage before indium-111 radiolabeling. In vitro studies of In-L-LDL particles show that indium labeling is rapidly achieved (1 h). More than 85% of the indium atoms are bound to the chelating functions of the incorporated DTPA derivatives and less than 10% to the nonspecific complexation sites of LDL (e.g., protein residues). After incubation in human serum, the indium activity recovered in the LDL fraction of In-L-LDL samples (95%) is much higher than in In-LDL samples (35%), pointing out the strong stabilizing chelating effect of the ligand. Competitive binding studies show that In-L-LDL are recognized by LDL receptors of A549 cells like native LDL when the In-L/LDL ratio varies from 5 to 30. All these in vitro experiments demonstrate that the In-L-LDL conjugates possess properties suitable for further work with in vivo experiments.
Collapse
Affiliation(s)
- F Jasanada
- Faculté de Pharmacie, Université Paul Sabatier, Toulouse, France
| | | | | | | | | | | |
Collapse
|
28
|
Notarnicola M, Linsalata M, Caruso MG, Cavallini A, Di Leo A. Low density lipoprotein receptors and polyamine levels in human colorectal adenocarcinoma. J Gastroenterol 1995; 30:705-9. [PMID: 8963386 DOI: 10.1007/bf02349635] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The low density lipoprotein receptor (LDLR) is a cell surface protein that binds with LDL, providing the cell with cholesterol for new membrane synthesis. Rapidly growing cells have high numbers of LDLRs, and these proteins have also been detected in neoplastic samples of human colorectal mucosa. Polyamines, putrescine, spermidine, and spermine, play an important role in cellular growth, and studies on colorectal cancers have demonstrated higher polyamine levels in neoplastic mucosa samples than in surrounding mucosa. The aim of this study was to investigate LDLR and polyamine levels in the neoplastic tissue of 43 patients (28 males and 15 females) with colorectal adenocarcinoma, using enzymatic immunoassay and high performance liquid chromatography, respectively. Specimens of neoplastic mucosa were considered LDLR-positive or LDLR-negative when the amount of bound human anti-LDLR antibody detected was equal or higher or lower than the cut-off value (0.5 ng of bound anti-LDLR Ab/mg protein), respectively. Twenty-one subjects were LDLR-positive and 22 LDLR-negative. Polyamine levels (nmol/g tissue) were higher in LDLR-positive specimens; this increase was significant for total polyamines (P < 0.05). These findings, reporting the presence of increased polyamine content in LDLR-positive colorectal neoplastic specimens, suggest an association between LDLR levels and gastrointestinal neoplastic proliferative activity.
Collapse
Affiliation(s)
- M Notarnicola
- Scientific Institute for Digestive Diseases, I.R.C.C.S. "S. de Bellis", Castellana, Italy
| | | | | | | | | |
Collapse
|
29
|
Niendorf A, Nägele H, Gerding D, Meyer-Pannwitt U, Gebhardt A. Increased LDL receptor mRNA expression in colon cancer is correlated with a rise in plasma cholesterol levels after curative surgery. Int J Cancer 1995; 61:461-4. [PMID: 7759150 DOI: 10.1002/ijc.2910610405] [Citation(s) in RCA: 49] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
It is currently under debate whether the low serum cholesterol levels that are frequently observed in cancer patients represent a risk factor for/or, rather, are a consequence of the tumour. We postulate that malignant tumours are directly involved in an increased catabolism of cholesterol-rich low-density lipoprotein (LDL) particles. In a prospective study of 25 patients with colorectal carcinoma, we measured intraindividual shifts in serum cholesterol levels after surgery, and the expression of LDL-receptor mRNA in surgically removed specimens. A significant rise in plasma cholesterol levels was observed in patients 3 and 12 months after curative surgery, but not after non-curative surgery. In human colon carcinoma tissues LDL receptor mRNA expression, as determined by competitive reverse-transcriptase-polymerase-chain reaction, was found to be significantly increased when compared to tissues from the tumour-free margin (median values, 1.2 x 10(6) vs. 2.0 x 10(5) molecules/micrograms total cellular RNA, respectively, n = 17). The extent of LDL-receptor mRNA expression positively correlated to the percentage rise of plasma cholesterol levels 3 months (n = 7, r = 0.8763) and 12 months (n = 6, r = 0.9181) after curative surgery. This finding provides in vivo evidence that the tumour tissue itself contributes to decreased plasma cholesterol levels in patients suffering from colorectal carcinomas. It supports the hypothesis that low cholesterol levels in cancer patients are a consequence, and not the cause, of the malignancy.
Collapse
Affiliation(s)
- A Niendorf
- Institute of Pathology, University of Hamburg, Germany
| | | | | | | | | |
Collapse
|
30
|
Windler E, Ewers-Grabow U, Thiery J, Walli A, Seidel D, Greten H. The prognostic value of hypocholesterolemia in hospitalized patients. THE CLINICAL INVESTIGATOR 1994; 72:939-43. [PMID: 7711423 DOI: 10.1007/bf00577732] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Clinical observations show that severe illness often leads to hypocholesterolemia. To verify this finding and to define the relationship between serum cholesterol and a patient's prognosis, a study was conducted in two large hospital populations. Of 24,000 and 61,463 adult patients (populations I and II) an average of 3.8% and 3.6% died in hospital, respectively. The mean serum cholesterol levels of patients who died was significantly lower than that of those who survived (163.6 mg/dl versus 217.8 mg/dl; P < 0.0001). The average cholesterol of surviving patients was similar to that of 6,543 healthy controls. During hospitalization serum cholesterol levels of < or = 100 mg/dl were encountered in 1.2% and 3.6% of patients of populations I and II, respectively. The mortality of these hypocholesterolemic patients was about tenfold higher than average and showed a strong, inverse, linear relationship with serum cholesterol concentrations. Patients whose serum cholesterol level dropped to less than 45 mg/dl did not survive. These data show that in severely ill patients serum cholesterol may decline to very low concentrations, and the prognosis is reflected by the degree of hypocholesterolemia, which thus may serve as a clinically useful prognostic parameter.
Collapse
Affiliation(s)
- E Windler
- Medizinische Kernklinik und Poliklinik, Universität-Krankenhaus Eppendorf, Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
31
|
Ponty E, Favre G, Benaniba R, Boneu A, Lucot H, Carton M, Soula G. Biodistribution study of 99mTc-labeled LDL in B16-melanoma-bearing mice. Visualization of a preferential uptake by the tumor. Int J Cancer 1993; 54:411-7. [PMID: 8509217 DOI: 10.1002/ijc.2910540311] [Citation(s) in RCA: 29] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Since there is strong evidence of a preferential LDL accumulation in tumor cells, LDL might be of interest for tumor imaging. We have tested the ability of 99mTc-LDL in tumor imaging with B16-melanoma-bearing mice as a model for further applications in human studies. The LDL fixation rate was higher with 99mTc-labeled LDL than with 125I labeled LDL. Since technetium-99m remains trapped in the cells, 99mTc-LDL is a well-adapted radioligand because of information given by this radiotracer on the receptor metabolism. We observed that, at early growth stages, the tumor took up the LDL at a maximal rate, suggesting differences in cholesterol metabolism as a function of tumor growth. Accumulation of label in the tumor area was perfectly observable in tumor-bearing mice on scintigraphic images. Computerized quantification of the regions of interest (as well as biodistribution studies including killing of the animals) showed a 1.81-fold increase in uptake by the tumor as compared to the liver and a 28-fold increase as compared with corresponding normal tissue (muscle of the left leg) at day 8 of tumor growth. These data give strong support to the value of this non-invasive method in visualizing and quantifying the tissue LDL uptake in vivo, including the precise information provided by nuclear scintigraphy on the distribution of the radiolabeled LDL in the different tissues. 99mTc-LDL could be an efficient tool for further diagnostic or therapeutic exploration in cancer patients.
Collapse
Affiliation(s)
- E Ponty
- Laboratoire de Ciblage en Thérapeutique, UFR des Sciences Pharmaceutiques, Toulouse, France
| | | | | | | | | | | | | |
Collapse
|
32
|
Törnberg S, Carstensen J. Serum beta-lipoprotein, serum cholesterol and Quetelet's index as predictors for survival of breast cancer patients. Eur J Cancer 1993; 29A:2025-30. [PMID: 8280497 DOI: 10.1016/0959-8049(93)90466-s] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
We studied the survival of breast cancer patients in relation to serum cholesterol level, serum beta-lipoprotein level (BLP) and being overweight among women having breast cancer diagnosed during a follow-up period of 20 years. A cohort of 46,570 women attended a general health screening including examination of serum lipid levels, height and weight during 1963-1965. Of these, 1170 women developing breast cancer; 196 were below the age of 50 and 974 were above 50 years of age. 66 of the younger women, and 341 of the older women were reported to have died of breast cancer. A correlation was found between high serum BLP and decreased survival of breast cancer patients < 50 years of age. For women > or = 60 years of age, BLP was positively correlated to breast cancer survival. No correlation was found between serum cholesterol level and breast cancer survival in any age group. Increasing obesity was statistically significantly correlated to decreased survival with breast cancer. The latter findings were in accordance with other studies which have shown being overweight as a risk factor for breast cancer. As for the relationships between ischaemic heart disease and serum lipid levels, in studies of cancer risks in relation to serum cholesterol level, the different fractions of cholesterol seem to be of importance.
Collapse
Affiliation(s)
- S Törnberg
- Department of General Oncology, Radiumhemmet, Karolinska Hospital, Stockholm, Sweden
| | | |
Collapse
|
33
|
Gueddari N, Favre G, Hachem H, Marek E, Le Gaillard F, Soula G. Evidence for up-regulated low density lipoprotein receptor in human lung adenocarcinoma cell line A549. Biochimie 1993; 75:811-9. [PMID: 8274533 DOI: 10.1016/0300-9084(93)90132-c] [Citation(s) in RCA: 56] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Human lung adenocarcinoma cell line A549 was studied with respect to the metabolism of human low density lipoprotein (LDL) and 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGR) activity. After incubation in medium containing lipoprotein-deficient serum (LPDS) for 24 h, the A549 cell line expresses a single class of high affinity LDL binding sites (KD at 37 degrees C of 15.1 +/- 0.7 nM and capacity of 118 +/- 2.8 ng/mg cell protein) and an HMGR activity of 111.4 +/- 7 pmol/min/mg cell protein. After binding, the LDLs were internalized and degraded by a common saturable process. The HMGR activity was higher in A549 cells than in fibroblasts but LDL affinity and binding capacity were similar in both cell types. However, in the presence of lipoproteins, A549 cells showed a two-fold higher binding capacity than fibroblasts. When the cells were deprived of cholesterol, the amount of LDLR sites increased but the extent of stimulation was lower in A549 than in fibroblast cells (2.5-fold versus six-fold respectively). This increase was accompanied by a similar increase in the specific LDLR mRNA cellular levels (two-fold versus six-fold respectively). When cells were deprived of exogenous and endogenous cholesterol (biosynthesis blocked by compactin), the binding capacity and the LDLR mRNA levels were yet again increased in A549 cells but not in fibroblasts. Taken together these results suggest that the level of expression of the LDLR is up-regulated in A549 cells compared to fibroblasts.
Collapse
Affiliation(s)
- N Gueddari
- Laboratoire de Ciblage en Thérapeutique, Faculté des Sciences Pharmaceutiques, Toulouse, France
| | | | | | | | | | | |
Collapse
|
34
|
Törnberg SA. Serum cholesterol levels and cancer. A research dilemma. Ann Med 1991; 23:93-4. [PMID: 2069798 DOI: 10.3109/07853899109148030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
|
35
|
Niendorf A, Beisiegel U. Low-density lipoprotein receptors. CURRENT TOPICS IN PATHOLOGY. ERGEBNISSE DER PATHOLOGIE 1991; 83:187-218. [PMID: 2007335 DOI: 10.1007/978-3-642-75515-6_8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
36
|
Rudling MJ, Reihnér E, Einarsson K, Ewerth S, Angelin B. Low density lipoprotein receptor-binding activity in human tissues: quantitative importance of hepatic receptors and evidence for regulation of their expression in vivo. Proc Natl Acad Sci U S A 1990; 87:3469-73. [PMID: 2110363 PMCID: PMC53922 DOI: 10.1073/pnas.87.9.3469] [Citation(s) in RCA: 76] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The heparin-sensitive binding of 125I-labeled low-density lipoprotein (LDL) to homogenates from 18 different normal human tissues and some solid tumors was determined. The binding to adrenal and liver homogenates fulfilled criteria established for the binding of LDL to its receptor--namely, (i) saturability, (ii) sensitivity to proteolytic destruction, (iii) inhibition by EDTA, and (iv) heat sensitivity. When the binding of 125I-labeled LDL was assayed at a constant concentration (50 micrograms/ml), the adrenal gland and the ovary had the highest binding of normal tissues. The highest binding per g of tissue overall was obtained in homogenates of a gastric carcinoma and a parotid adenoma. When the weights of the parenchymatous organs were considered, the major amount of LDL receptors was contained in the liver. To study the possible regulation of hepatic LDL-receptor expression, 11 patients were pretreated with cholestyramine (8 g twice a day for 3 weeks). Increased binding activity (+105%, P less than 0.001) was obtained in homogenates from liver biopsies from the cholestyramine-treated patients as compared with 12 untreated controls. It is concluded that the liver is the most important organ for LDL catabolism in humans and that the receptor activity in this organ can be regulated upon pharmacologic intervention. Further studies are needed to confirm the possibility that certain solid tumors can exhibit high numbers of LDL receptors.
Collapse
Affiliation(s)
- M J Rudling
- Department of Internal Medicine, Karolinska Institutet at Huddinge University Hospital, Sweden
| | | | | | | | | |
Collapse
|
37
|
Henriksson P, Eriksson M, Ericsson S, Rudling M, Stege R, Berglund L, Angelin B. Hypocholesterolaemia and increased elimination of low-density lipoproteins in metastatic cancer of the prostate. Lancet 1989; 2:1178-80. [PMID: 2572901 DOI: 10.1016/s0140-6736(89)91790-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
To study the influence of tumour mass on lipid metabolism, the lipoprotein pattern in untreated patients with newly diagnosed cancer of the prostate was examined. Total cholesterol levels were reduced in patients with evidence of metastasis (n = 30) compared with those without metastasis (n = 73). Since the major fraction of serum cholesterol is contained in low-density lipoproteins (LDL), turnover of LDL was studied in detail in 8 patients compared with 12 age-matched healthy men. LDL were cleared faster in the 3 patients with metastatic disease than in the patients without metastasis and in controls, indicating faster catabolism of LDL. Thus in prostatic cancer an increased tumour burden is associated with increased elimination of LDL, which contributes to reduced serum cholesterol levels.
Collapse
Affiliation(s)
- P Henriksson
- Department of Medicine, Karolinska Institute, Huddinge University Hospital, Sweden
| | | | | | | | | | | | | |
Collapse
|
38
|
Törnberg SA, Holm LE, Carstensen JM. Breast cancer risk in relation to serum cholesterol, serum beta-lipoprotein, height, weight, and blood pressure. Acta Oncol 1988; 27:31-7. [PMID: 3365352 DOI: 10.3109/02841868809090315] [Citation(s) in RCA: 98] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
The relation between breast cancer risk and serum levels of cholesterol and beta-lipoprotein (BLP), height, weight, Quetelet's index and blood pressure was studied in a cohort of 46,570 Swedish women less than 75 years of age. The cohort was examined between 1963 and 1965 and followed up in the Swedish Cancer Registry until 1983. During this period 1,182 cases of breast cancer were reported. Of those, 196 were reported among women less than 50 years of age. Statistically significant positive associations were observed between height, weight, and systolic blood pressure and breast cancer risk. No clear trend in cancer risk related to serum cholesterol or BLP was seen in the total material. In a stepwise Cox multiple regression analysis only the associations with height and blood pressure remained significant. Among women, having their cancer diagnosed before the age of 50, higher Quetelet's index was associated with a lower cancer risk, whereas a positive correlation was seen among women greater than or equal to 50 years. In the group of younger women a high BLP level was associated with an increased risk of breast cancer. This relation became even stronger when studied in a multivariate analysis, which also showed a negative correlation between serum cholesterol and cancer risk.
Collapse
Affiliation(s)
- S A Törnberg
- Department of General Oncology, Karolinska Sjukhuset, Stockholm, Sweden
| | | | | |
Collapse
|