1
|
Frey C. A Real-World Pharmacovigilance Analysis of Lorlatinib-Associated Metabolic Effects Using The FDA Adverse Events Reporting System (FAERS) Database From 2013 to 2024. Endocr Pract 2025:S1530-891X(25)00132-6. [PMID: 40287137 DOI: 10.1016/j.eprac.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/29/2025]
Abstract
OBJECTIVE The advent of anaplastic lymphoma kinase (ALK) inhibitors, including lorlatinib, has transformed the treatment of ALK-rearranged malignancies. While lorlatinib effectively overcomes resistance mutations and penetrates the central nervous system, its use is associated with metabolic adverse events, including hypercholesterolemia, hypertriglyceridemia, and weight gain. These complications increase cardiovascular risks, disrupt metabolic homeostasis, and may affect therapy adherence. METHODS This study utilizes data from the FDA Adverse Event Reporting System (FAERS) and employs disproportionality analysis to investigate the prevalence and nature of lorlatinib-associated metabolic adverse events. RESULTS Significant associations were identified between lorlatinib and lipid-related adverse events, including hypercholesterolemia (ROR = 98.46; 95% CI: 79.28-122.29), hypertriglyceridemia (ROR = 66.10; 95% CI: 49.60-88.11), increased body mass index (ROR = 81.57; 95% CI: 48.87-136.14), and increased blood cholesterol (ROR = 23.42; 95% CI: 19.69-27.86). Additional associations were noted for increased blood triglycerides (ROR = 28.14; 95% CI: 22.15-35.75) and dyslipidemia (ROR = 53.60; 95% CI: 38.51-74.60). CONCLUSION These findings highlight the need for proactive monitoring and management of metabolic side effects in patients receiving lorlatinib. A multidisciplinary approach-incorporating pharmacological interventions, lifestyle modifications, and regular monitoring-is essential to mitigate metabolic risks. This study enhances the understanding of lorlatinib's safety profile and informs clinical strategies to balance efficacy and tolerability in ALK inhibitor therapy.
Collapse
Affiliation(s)
- Connor Frey
- Department of Medicine, University of British Columbia, 317 - 2194 Health Sciences Mall, Vancouver, British Columbia, V6T 1Z3, Canada
| |
Collapse
|
2
|
Mazzilli R, Zamponi V, Mancini C, Giorgini B, Golisano B, Mikovic N, Pecora G, Russo F, Martiradonna M, Paravani P, Prosperi D, Faggiano A. Neuroendocrine tumors and diabetes mellitus: which treatment and which effect. Endocrine 2025; 88:36-50. [PMID: 39752043 DOI: 10.1007/s12020-024-04149-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/23/2024] [Indexed: 01/04/2025]
Abstract
Diabetes mellitus (DM) and neuroendocrine tumors (NET) can exert unfavorable effects on each other prognosis. In this narrative review, we evaluated the effects of NET therapies on glycemic control and DM management and the effects of anti-diabetic therapies on NET outcome and management. For this purpose, we searched the PubMed, Science Direct, and Google Scholar databases for studies reporting the effects of NET therapy on DM as well as the effect of DM therapy on NET. The majority of NET treatments appear to impair glycaemic control, both inducing hypoglycemic or, more commonly, hyperglycemia and even new-onset DM. However, glucose metabolism imbalance can be effectively managed by modulating anti-diabetic therapy and adopting an appropriate nutritional approach. On the other hand, the effects of anti-diabetic treatment, like insulin, sulfonylureas, thiazolidinediones, ipeptidyl-peptidase-4 inhibitors, Glucagon-like peptide-1 receptor agonists, and Sodium-glucose cotransporter-2 inhibitors on NET are unclear. Recently, metformin has been investigated in patients with gastroenteropancreatic NET resulting in improved progression free survival suggesting a potential antineoplastic role. Finally, the management of DM in patients with NET is of great clinical relevance to correctly perform radiological procedures and even more functional imaging procedures, as well as to optimize the therapy and avoid treatment withdrawal or discontinuation. In conclusion, understanding the mechanisms underlying therapy-induced DM and implementing appropriate monitoring and management strategies of DM are essential for optimizing NET patient outcome and quality of life.
Collapse
Affiliation(s)
- Rossella Mazzilli
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy.
| | - Virginia Zamponi
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| | - Camilla Mancini
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Beatrice Giorgini
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| | - Bianca Golisano
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| | - Nevena Mikovic
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| | - Giulia Pecora
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| | - Flaminia Russo
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| | - Maurizio Martiradonna
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| | - Piero Paravani
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| | - Daniela Prosperi
- Unit of Nuclear Medicine, ENETS Center of Excellence, Sant'Andrea University Hospital, Rome, Italy
| | - Antongiulio Faggiano
- Unit of Endocrinology, Department of Clinical and Molecular Medicine, ENETS Center of Excellence, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
3
|
Geetha AVS, Harithpriya K, Ganesan K, Ramkumar KM. Exploring the Role of Hypoxia and HIF-1α in the Intersection of Type 2 Diabetes Mellitus and Endometrial Cancer. Curr Oncol 2025; 32:106. [PMID: 39996906 PMCID: PMC11854729 DOI: 10.3390/curroncol32020106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/26/2025] Open
Abstract
Diabetes and Cancer are the most complex chronic diseases, accounting for significant global mortality and morbidity. The association between Type 2 DM (T2DM) and endometrial cancer (EC) is multifaced, sharing numerous risk factors, including insulin resistance, obesity, hypoxia, and oxidative stress. Hypoxia plays a vital role in T2DM pathogenesis by altering the insulin level and pancreatic β-cell failure through an imbalance between antioxidant enzymes and cellular oxidative levels, while chronic inflammation contributes to EC malignancy. HIF-1α is a potent transcription factor involved in modulating cellular responses to hypoxia within the disease environment. Targeting the HIF-1α signaling cascade, a major metabolic regulator may contribute to advanced therapeutic advances. This review focuses on the association between T2DM and EC, especially focusing on hypoxia and HIF signaling pathways. These intersect with key pathways involved in T2DM and EC pathology, such as insulin signaling, PI3K/AKT, mTOR pathway, MUC1/HIF-1α pathway, and hormonal imbalance. Understanding this complex relationship paves the way for future researchers to develop HIF-1α-targeted therapies that could lead to novel combination therapies to treat these comorbid conditions.
Collapse
Affiliation(s)
- Alagappan V. S. Geetha
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, India; (A.V.S.G.); (K.H.)
| | - Kannan Harithpriya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, India; (A.V.S.G.); (K.H.)
| | - Kumar Ganesan
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China;
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, India; (A.V.S.G.); (K.H.)
| |
Collapse
|
4
|
Fan K, Wang Y, Bian J, Sun Y, Dou J, Pan J, Yu Y. Study on the effects of rapamycin and the mTORC1/2 dual inhibitor OSI-027 on the metabolism of colon cancer cells based on UPLC-MS/MS metabolomics. Invest New Drugs 2024; 42:418-427. [PMID: 38916794 DOI: 10.1007/s10637-024-01438-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 04/16/2024] [Indexed: 06/26/2024]
Abstract
mTORC1/2 dual inhibitors may be more effective than mTORC1 inhibitor rapamycin. However, their metabolic impacts on colon cancer cells remain unexplored. We conducted a comparative analysis of the anti-proliferative effects of rapamycin and the novel OSI-027 in colon cancer cells HCT-116, evaluating their metabolic influences through ultra-high performance liquid chromatography-mass spectrometry (UPLC-MS/MS). Our results demonstrate that OSI-027 more effectively inhibits colon cancer cell proliferation than rapamycin. Additionally, we identified nearly 600 metabolites from the spectra, revealing significant differences in metabolic patterns between cells treated with OSI-027 and rapamycin. Through VIP value screening, we pinpointed crucial metabolites contributing to these distinctions. For inhibiting glycolysis and reducing glucose consumption, OSI-027 was likely to be more potent than rapamycin. For amino acids metabolism, although OSI-027 has a broad effect as rapamycin, their effects in degrees were not exactly the same. These findings address the knowledge gap regarding mTORC1/2 dual inhibitors and lay a foundation for their further development and research.
Collapse
Affiliation(s)
- Kai Fan
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, 1055 San Xiang Road, Suzhou City, 215004, Jiangsu, People's Republic of China
| | - Yueyuan Wang
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, 1055 San Xiang Road, Suzhou City, 215004, Jiangsu, People's Republic of China
| | - Jiangyujing Bian
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, 1055 San Xiang Road, Suzhou City, 215004, Jiangsu, People's Republic of China
- College of Pharmacy, Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Yewen Sun
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, 1055 San Xiang Road, Suzhou City, 215004, Jiangsu, People's Republic of China
- College of Pharmacy, Soochow University, Suzhou, Jiangsu, People's Republic of China
| | - Jiaqi Dou
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, 1055 San Xiang Road, Suzhou City, 215004, Jiangsu, People's Republic of China
| | - Jie Pan
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, 1055 San Xiang Road, Suzhou City, 215004, Jiangsu, People's Republic of China.
| | - Yunli Yu
- Department of Pharmacy, The Second Affiliated Hospital of Soochow University, 1055 San Xiang Road, Suzhou City, 215004, Jiangsu, People's Republic of China.
| |
Collapse
|
5
|
Braegelmann J, Führer D, Tan S. [Endocrine side effects of tumor treatment]. INNERE MEDIZIN (HEIDELBERG, GERMANY) 2024; 65:681-689. [PMID: 38874811 DOI: 10.1007/s00108-024-01731-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/23/2024] [Indexed: 06/15/2024]
Abstract
Targeted and immune-based treatments represent significant innovations in oncology and impressively improve the prognosis of many tumor diseases. Their now widespread use as a standard treatment for several malignant diseases increasingly requires knowledge of how to deal with new adverse events (AE) induced by oncological agents in centers and routine practice [12, 13]. For example, the blockade of specific checkpoints of the inhibitory immune system by immune checkpoint inhibitors (ICI) causes the loss of immune tolerance to the body's own tissue with the occurrence of endocrine immune-related AE (irAE) in approximately 10% of patients treated with ICI [3, 11]. Targeted treatments, such as with tyrosine kinase inhibitors (TKI), mammalian target of rapamycin (mTOR) and phosphoinositide 3‑kinase (PI3K) inhibitors often lead to disorders of glucose metabolism and thyroid gland dysfunction. The challenges of maintaining bone health during endocrine therapy in patients with prostate and hormone receptor-positive breast cancer and in the endocrine follow-up care of childhood cancer survivors are well-known and are becoming increasingly more important for the long-term prognosis and quality of life [5, 20]. However, although the recommendations for a systematic management of endocrine side effects of these relatively new tumor therapies can be found in guidelines, they are not yet established in routine clinical care [15, 19]. A close interdisciplinary cooperation is required for optimal care of people with cancer [7]. The development of such interdisciplinary cross-sectoral treatment structures is important as tumor treatment is primarily carried out by hematologists or oncologists, while the management of AE induced by oncological agents increasingly involves primary care physicians including internists and in the case of endocrine AE requires the specific expertise of endocrinologists and diabetologists.
Collapse
Affiliation(s)
- Johanna Braegelmann
- Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Universitätsklinikum Essen, Universitätsmedizin Essen, Hufelandstraße 55, 45147, Essen, Deutschland
| | - Dagmar Führer
- Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Universitätsklinikum Essen, Universitätsmedizin Essen, Hufelandstraße 55, 45147, Essen, Deutschland
| | - Susanne Tan
- Klinik für Endokrinologie, Diabetologie und Stoffwechsel, Universitätsklinikum Essen, Universitätsmedizin Essen, Hufelandstraße 55, 45147, Essen, Deutschland.
| |
Collapse
|
6
|
Calderón Guzmán D, Osnaya Brizuela N, Ortíz Herrera M, Valenzuela Peraza A, Labra Ruíz N, Juárez Olguín H, Santamaria del Angel D, Barragán Mejía G. N-Acetylcysteine Attenuates Cisplatin Toxicity in the Cerebrum and Lung of Young Rats with Artificially Induced Protein Deficiency. Int J Mol Sci 2024; 25:6239. [PMID: 38892427 PMCID: PMC11172823 DOI: 10.3390/ijms25116239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/15/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Neurotoxicity is a major obstacle in the effectiveness of Cisplatin in cancer chemotherapy. In this process, oxidative stress and inflammation are considered to be the main mechanisms involved in brain and lung toxicity. The aim of the present work was to study the influence of the amount of protein on some oxidative parameters in the brain and lungs of rats treated with Cisplatin (CP) and N-Acetylcysteine (NAC) as neuroprotectors. Four groups of Wistar rats, each containing six animals, were fed with a protein diet at 7% for 15 days. Thereafter, the groups were given either a unique dose of CP® 5 mg/kg or NAC® 5 mg/kg as follows: group 1 (control), NaCl 0.9% vehicle; group 2, CP; group 3, NAC; and group 4, NAC + CP. The animals were sacrificed immediately after the treatments. Blood samples were collected upon sacrifice and used to measure blood triglycerides and glucose. The brain and lungs of each animal were obtained and used to assay lipid peroxidation (TBARS), glutathione (GSH), serotonin metabolite (5-HIAA), catalase, and the activity of Ca+2, and Mg+2 ATPase using validated methods. TBARS, H2O2, and GSH were found to be significantly decreased in the cortex and cerebellum/medulla oblongata of the groups treated with CP and NAC. The total ATPase showed a significant increase in the lung and cerebellum/medulla oblongata, while 5-HIAA showed the same tendency in the cortex of the same group of animals. The increase in 5-HIAA and ATPase during NAC and CP administration resulted in brain protection. This effect could be even more powerful when membrane fluidity is increased, thus proving the efficacy of combined NAC and CP drug therapy, which appears to be a promising strategy for future chemotherapy in malnourished patients.
Collapse
Affiliation(s)
- David Calderón Guzmán
- Laboratory of Neurosciences, Instituto Nacional de Pediatria (INP), Mexico City 04530, Mexico; (D.C.G.); (N.O.B.); (A.V.P.); (N.L.R.); (D.S.d.A.)
| | - Norma Osnaya Brizuela
- Laboratory of Neurosciences, Instituto Nacional de Pediatria (INP), Mexico City 04530, Mexico; (D.C.G.); (N.O.B.); (A.V.P.); (N.L.R.); (D.S.d.A.)
| | - Maribel Ortíz Herrera
- Laboratory of Experimental Bacteriology, Instituto Nacional de Pediatria INP, Mexico City 04530, Mexico; (M.O.H.); (G.B.M.)
| | - Armando Valenzuela Peraza
- Laboratory of Neurosciences, Instituto Nacional de Pediatria (INP), Mexico City 04530, Mexico; (D.C.G.); (N.O.B.); (A.V.P.); (N.L.R.); (D.S.d.A.)
| | - Norma Labra Ruíz
- Laboratory of Neurosciences, Instituto Nacional de Pediatria (INP), Mexico City 04530, Mexico; (D.C.G.); (N.O.B.); (A.V.P.); (N.L.R.); (D.S.d.A.)
| | - Hugo Juárez Olguín
- Laboratory of Pharmacology, Instituto Nacional de Pediatría, Avenida Imán N° 1, 3rd piso Colonia Cuicuilco, Mexico City 04530, Mexico
| | - Daniel Santamaria del Angel
- Laboratory of Neurosciences, Instituto Nacional de Pediatria (INP), Mexico City 04530, Mexico; (D.C.G.); (N.O.B.); (A.V.P.); (N.L.R.); (D.S.d.A.)
| | - Gerardo Barragán Mejía
- Laboratory of Experimental Bacteriology, Instituto Nacional de Pediatria INP, Mexico City 04530, Mexico; (M.O.H.); (G.B.M.)
| |
Collapse
|
7
|
Abdulwadood I, De melo JF, Scheel RC, Bois JP. Severe Hypertriglyceridemia in a Patient With Metabolic Syndrome and Psoriasis on Risankizumab-Rzaa. JCEM CASE REPORTS 2024; 2:luae087. [PMID: 38911361 PMCID: PMC11191645 DOI: 10.1210/jcemcr/luae087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Indexed: 06/25/2024]
Abstract
We report a case of severe hypertriglyceridemia (HTG) complicated by hyperviscosity syndrome as a possible adverse reaction to risankizumab-rzaa in a 49-year-old male with a history of longstanding uncontrolled type 2 diabetes, obesity, and coronary artery disease with prior ST-elevation myocardial infarction. On admission, the patient presented with xanthomatous plaques, chest and epigastric discomfort, and headache. Subsequent blood testing revealed severely elevated triglyceride (TG) levels at 7670 mg/dL (86.59 mmol/L) [reference range: <150 mg/dL; 1.69 mmol/L] and total cholesterol at 934 mg/dL (24.14 mmol/L) [reference range: <200 mg/dL; 5.17 mmol/L]. Triglyceride levels decreased and symptoms resolved with dietary restrictions and plasmapheresis. At follow-up, his TG remained elevated but improved, and he was advised to continue lipid-lowering medications as well as cessation of risankizumab. While the patient presented with high risk factors, we posit that the subacute presentation of severe HTG is a possible result of his recent course of risankizumab-rzaa therapy for management of psoriasis. This is noteworthy as pharmaceutical surveys and clinical trials do not list severe HTG as an adverse effect. Postmarketing surveillance studies are essential to confirm this potential association and monitor drug safety. In summary, this case highlights a possible link between risankizumab and severe HTG, emphasizing the importance of ongoing pharmacovigilance to identify and manage unexpected adverse effects associated with new medications.
Collapse
Affiliation(s)
| | - Jose F De melo
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Robert C Scheel
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - John P Bois
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
8
|
Silvestris N, Franchina T, Gallo M, Argentiero A, Avogaro A, Cirino G, Colao A, Danesi R, Di Cianni G, D'Oronzo S, Faggiano A, Fogli S, Giuffrida D, Gori S, Marrano N, Mazzilli R, Monami M, Montagnani M, Morviducci L, Natalicchio A, Ragni A, Renzelli V, Russo A, Sciacca L, Tuveri E, Zatelli MC, Giorgino F, Cinieri S. Diabetes management in cancer patients. An Italian Association of Medical Oncology, Italian Association of Medical Diabetologists, Italian Society of Diabetology, Italian Society of Endocrinology and Italian Society of Pharmacology multidisciplinary consensus position paper. ESMO Open 2023; 8:102062. [PMID: 38070434 PMCID: PMC10714217 DOI: 10.1016/j.esmoop.2023.102062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/07/2023] [Accepted: 10/11/2023] [Indexed: 12/31/2023] Open
Abstract
Cancer management has significantly evolved in recent years, focusing on a multidisciplinary team approach to provide the best possible patient care and address the various comorbidities, toxicities, and complications that may arise during the patient's treatment journey. The co-occurrence of diabetes and cancer presents a significant challenge for health care professionals worldwide. Management of these conditions requires a holistic approach to improve patients' overall health, treatment outcomes, and quality of life, preventing diabetes complications and cancer treatment side-effects. In this article, a multidisciplinary panel of experts from different Italian scientific societies provide a critical overview of the co-management of cancer and diabetes, with an increasing focus on identifying a novel specialty field, 'diabeto-oncology', and suggest new co-management models of cancer patients with diabetes to improve their care. To better support cancer patients with diabetes and ensure high levels of coordinated care between oncologists and diabetologists, 'diabeto-oncology' could represent a new specialized field that combines specific expertise, skills, and training.
Collapse
Affiliation(s)
- N Silvestris
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina.
| | - T Franchina
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina
| | - M Gallo
- Endocrinology and Metabolic Diseases Unit, AO SS Antonio e Biagio e Cesare Arrigo of Alessandria, Alessandria
| | - A Argentiero
- Medical Oncology Unit, IRCCS Istituto Tumori "Giovanni Paolo II", Bari
| | - A Avogaro
- Department of Medicine, University of Padova, Padua
| | - G Cirino
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples
| | - A Colao
- Endocrinology, Diabetology and Andrology Unit, Department of Clinical Medicine and Surgery, Federico II University of Naples, Naples; UNESCO Chair, Education for Health and Sustainable Development, Federico II University, Naples
| | - R Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa
| | | | - S D'Oronzo
- Interdisciplinary Department of Medicine, University of Bari Aldo Moro, Bari
| | - A Faggiano
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, Rome
| | - S Fogli
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Pisa
| | - D Giuffrida
- Department of Oncology, Istituto Oncologico del Mediterraneo, Viagrande, Catania
| | - S Gori
- Oncologia Medica, IRCCS Ospedale Don Calabria-Sacro Cuore di Negrar, Verona
| | - N Marrano
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari
| | - R Mazzilli
- Endocrinology Unit, Department of Clinical and Molecular Medicine, Sant'Andrea Hospital, ENETS Center of Excellence, Sapienza University of Rome, Rome
| | - M Monami
- Diabetology, Careggi Hospital and University of Florence, Firenze
| | - M Montagnani
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Pharmacology, Medical School, University of Bari Aldo Moro, Bari
| | - L Morviducci
- Diabetology and Nutrition Unit, Department of Medical Specialties, ASL Roma 1 - S, Spirito Hospital, Rome
| | - A Natalicchio
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari
| | - A Ragni
- Endocrinology and Metabolic Diseases Unit, AO SS Antonio e Biagio e Cesare Arrigo of Alessandria, Alessandria
| | - V Renzelli
- Diabetologist and Endocrinologist, Italian Association of Medical Diabetologists, Rome
| | - A Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo
| | - L Sciacca
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania, Catania
| | - E Tuveri
- Diabetology, Endocrinology and Metabolic Diseases Service, ASL-Sulcis, Carbonia
| | - M C Zatelli
- Section of Endocrinology, Geriatrics, and Internal Medicine, Department of Medical Sciences, University of Ferrara, Ferrara
| | - F Giorgino
- Department of Precision and Regenerative Medicine and Ionian Area, Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, University of Bari Aldo Moro, Bari
| | - S Cinieri
- Medical Oncology Division and Breast Unit, Senatore Antonio Perrino Hospital, ASL Brindisi, Brindisi, Italy
| |
Collapse
|
9
|
Zhu Z, Xu S, Ren J, Jiang T, Zhang C, Yan Z. Anlotinib affects systemic lipid metabolism and induces lipid accumulation in human lung cancer cells. Lipids Health Dis 2023; 22:134. [PMID: 37612751 PMCID: PMC10464365 DOI: 10.1186/s12944-023-01907-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 08/16/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Anlotinib has demonstrated encouraging clinical outcomes in the treatment of lung cancer, soft tissue sarcoma and thyroid carcinoma. Several clinical studies have shown a relationship between anlotinib treatment and the occurrence of hyperlipidemia. The fundamental mechanisms, however, are still largely unclear. Here, the effect of anlotinib on lipid metabolism in an animal model and human cancer cells was evaluated and the role of lipid metabolism in the antitumor efficacy of anlotinib was investigated. METHODS The C57BL/6 J mouse model as well as A549 and H460 human lung cancer cell lines were used to examine the impact of anlotinib on lipid metabolism both in vivo and in vitro. Levels of triglycerides, high-density lipoprotein, low-density lipoprotein (LDL), and total cholesterol in serum or cell samples were determined using assay kits. The expression levels of crucial genes and proteins involved in lipid metabolism were measured by quantitative RT-PCR and Western blotting. Furthermore, exogenous LDL and knockdown of low-density lipoprotein receptor (LDLR) were used in H460 cells to investigate the relevance of lipid metabolism in the anticancer efficacy of anlotinib. RESULTS Anlotinib caused hyperlipidemia in C57BL/6 J mice, possibly by downregulating hepatic LDLR-mediated uptake of LDL cholesterol. AMP-activated protein kinase and mammalian target of rapamycin inhibition may also be involved. Additionally, anlotinib enhanced sterol response element binding protein 1/2 nuclear accumulation as well as upregulated LDLR expression in A549 and H460 cells, which may be attributable to intracellular lipid accumulation. Knockdown of LDLR reduced intracellular cholesterol content, but interestingly, anlotinib significantly improved intracellular cholesterol accumulation in LDLR-knockdown cells. Both exogenous LDL and LDLR knockdown decreased the sensitivity of cells to anlotinib. CONCLUSIONS Anlotinib modulates host lipid metabolism through multiple pathways. Anlotinib also exerts a significant impact on lipid metabolism in cancer cells by regulating key transcription factors and metabolic enzymes. In addition, these findings suggest lipid metabolism is implicated in anlotinib sensitivity.
Collapse
Affiliation(s)
- Zhongling Zhu
- Department of Clinical Pharmacology, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Shan Xu
- Department of Clinical Pharmacology, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Jing Ren
- Department of Clinical Pharmacology, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Teng Jiang
- Department of Clinical Pharmacology, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Cai Zhang
- Department of Clinical Pharmacology, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
| | - Zhao Yan
- Department of Clinical Pharmacology, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China.
- Department of Continuing Education and Science and Technology Service, China Anti-Cancer Association, Tianjin, China.
| |
Collapse
|
10
|
Wan Q, Huang B, Li T, Xiao Y, He Y, Du W, Wang BZ, Dakin GF, Rosenbaum M, Goncalves MD, Chen S, Leong KW, Qiang L. Selective targeting of visceral adiposity by polycation nanomedicine. NATURE NANOTECHNOLOGY 2022; 17:1311-1321. [PMID: 36456644 DOI: 10.1038/s41565-022-01249-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 10/03/2022] [Indexed: 06/17/2023]
Abstract
Obesity is a pandemic health problem with poor solutions, especially for targeted treatment. Here we develop a polycation-based nanomedicine polyamidoamine generation 3 (P-G3) that-when delivered intraperitoneally-selectively targets visceral fat due to its high charge density. Moreover, P-G3 treatment of obese mice inhibits visceral adiposity, increases energy expenditure, prevents obesity and alleviates the associated metabolic dysfunctions. In vitro adipogenesis models and single-cell RNA sequencing revealed that P-G3 uncouples adipocyte lipid synthesis and storage from adipocyte development to create adipocytes that possess normal functions but are deficient in hypertrophic growth, at least through synergistically modulating nutrient-sensing signalling pathways. The visceral fat distribution of P-G3 is enhanced by modifying P-G3 with cholesterol to form lipophilic nanoparticles, which is effective in treating obesity. Our study highlights a strategy to target visceral adiposity and suggests that cationic nanomaterials could be exploited for treating metabolic diseases.
Collapse
Affiliation(s)
- Qianfen Wan
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Baoding Huang
- Department of Orthopaedic Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University and Guangdong Provincial Key Laboratory of Orthopaedics and Traumatology, Guangzhou, China
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Tianyu Li
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Yang Xiao
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Ying He
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Wen Du
- Department of Medicine, Columbia University, New York, NY, USA
| | - Branden Z Wang
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Gregory F Dakin
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Michael Rosenbaum
- Department of Medicine, Columbia University, New York, NY, USA
- Department of Pediatrics, Columbia University, New York, NY, USA
| | | | - Shuibing Chen
- Department of Surgery, Weill Cornell Medicine, New York, NY, USA
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.
| | - Li Qiang
- Naomi Berrie Diabetes Center and Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
11
|
Real-World Experience of Monitoring Practice of Endocrinopathies Associated with the Use of Novel Targeted Therapies among Patients with Solid Tumors. MEDICAL SCIENCES (BASEL, SWITZERLAND) 2022; 10:medsci10040065. [PMID: 36412906 PMCID: PMC9680233 DOI: 10.3390/medsci10040065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/16/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND Cancer treatments have gradually evolved into targeted molecular therapies characterized by a unique mechanism of action instead of non-specific cytotoxic chemotherapies. However, they have unique safety concerns. For instance, endocrinopathies, which are defined as unfavorable metabolic alterations including thyroid disorders, hyperglycemia, dyslipidemia, and adrenal insufficiency necessitate additional monitoring. The aim of this study was to assess the prevalence of monitoring errors and develop strategies for monitoring cancer patients who receive targeted therapies. METHOD A retrospective chart review was used to assess the prevalence of monitoring errors of endocrinopathies among cancer patients who received targeted therapies over one year. All of the adult cancer patients diagnosed with a solid tumor who received targeted therapies were included. The primary outcome was to determine the prevalence of monitoring errors of endocrinopathies. The secondary outcomes were to assess the incidences of endocrinopathies and referral practice to endocrinology services. RESULTS A total of 128 adult patients with solid tumors were involved. The primary outcome revealed a total of 148 monitoring errors of endocrinopathies. Monitoring errors of the lipid profile and thyroid functions were the most common error types in 94% and 92.6% of the patients treated with novel targeted therapies, respectively. Subsequently, 57% of the monitoring errors in the blood glucose measures were identified. Targeted therapies caused 63 events of endocrinopathies, hyperglycemia in 32% of the patients, thyroid disorders in 15.6% of them and dyslipidemia in 1.5% of the patients. CONCLUSION Our study showed a high prevalence of monitoring errors among the cancer patients who received targeted therapies which led to endocrinopathies. It emphasizes the importance of adhering to monitoring strategies and following up on the appropriate referral process.
Collapse
|
12
|
Ni Z, Xu S, Yu Z, Ye Z, Li R, Chen C, Yang J, Liu H, Zhou Z, Zhang X. Comparison of dual mTORC1/2 inhibitor AZD8055 and mTORC1 inhibitor rapamycin on the metabolism of breast cancer cells using proton nuclear magnetic resonance spectroscopy metabolomics. Invest New Drugs 2022; 40:1206-1215. [PMID: 36063263 DOI: 10.1007/s10637-022-01268-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 06/06/2022] [Indexed: 11/28/2022]
Abstract
Dual mTORC1/2 inhibitors may be more effective than mTORC1 inhibitor rapamycin. Nevertheless, their metabolic effects on breast cancer cells have not been reported. We compared the anti-proliferative capacity of rapamycin and a novel mTORC1/2 dual inhibitor (AZD8055) in two breast cancer cell lines (MDA-MB-231 and MDA-MB-453) and analyzed their metabolic effects using proton nuclear magnetic resonance (1H NMR) spectroscopy-based metabolomics. We found that AZD8055 more strongly inhibited breast cancer cell proliferation than rapamycin. The half-inhibitory concentration of AZD8055 in breast cancer cells was almost one-tenth that of rapamycin. We identified 22 and 23 metabolites from the 1H NMR spectra of MDA-MB-231 and MDA-MB-453 cells. The patterns of AZD8055- and rapamycin-treated breast cancer cells differed significantly; we then selected the metabolites that contributed to these differences. For inhibiting glycolysis and reducing glucose consumption, AZD8055 was likely to be more potent than rapamycin. For amino acids metabolism, although AZD8055 has a broad effect as rapamycin, their effects in degrees were not exactly the same. AZD8055 and rapamycin displayed cell-specific metabolic effects on breast cancer cells, a finding that deserves further study. These findings help fill the knowledge gap concerning dual mTORC1/2 inhibitors and provide a theoretical basis for their development.
Collapse
Affiliation(s)
- Zhitao Ni
- Clinical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shaolin Xu
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zheng Yu
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhongjiang Ye
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Rongqi Li
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chuang Chen
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jianhui Yang
- Department of Pharmacy, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Huamin Liu
- College of Pharmacy, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ziye Zhou
- Clinical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| | - Xiuhua Zhang
- Clinical Research Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
13
|
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: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [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
Abstract
Ligand-based targeting of the receptors that are overexpressed explicitly on cancer cells represents an effective drug delivery approach to enhance the chemotherapeutic efficacy. Proprotein convertase subtilisin/kexin type 9 (PCSK9) which is a serine protease enzyme primarily produced by the liver cells, can potentially be used as a targeting ligand. PCSK9 binds to the LDL-r on hepatocytes' surface, leading to endocytosis and endosomal degradation. High LDL-r expression, which is believed to meet the higher demand of the cholesterol and phospholipids to build proliferating cancer cell membrane, ensures selective uptake of the PCSK9 conjugated liposomes. In the present work, the PCSK9 conjugated liposomal system was developed to deliver paclitaxel (PTX) to cancer cells. The protein was conjugated by EDC and NHS in a two-step coupling reaction to the liposomes containing COOH-PEG2000-COOH lipid. Conjugation was confirmed by NMR, and liposomes were further characterized by SEM and zeta sizer. PCSK9-conjugated liposomes showed high encapsulation efficiency of 69.1% with a diameter of 90.0 ± 4.9 nm. Long-term stability (30 days) study (Zeta potential: -9.88) confirmed excellent constancy and significant drug retention (58.2%). Invitro cytotoxicity and targeting efficiency was explored using MTS assay in human embryonic kidney cells (HEK293), liver hepatocellular cells (HEPG2), and a human colon cancer cell line (HCT116) for 24 h. PCSK9 conjugated liposomes exhibited significantly higher growth inhibition than the unconjugated (control) liposomes in HCT116 cell line (p < 0.001). The novel PCSK9 conjugated liposomes presented potent and precise in vitro anticancer activity and, therefore, are suggested for the first time as a promising targeted delivery system for cancer treatment.
Collapse
Affiliation(s)
- Nitin Bharat Charbe
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Av. Vicuña McKenna 4860, 7820436, Macul, Santiago, Chile; Department of Pharmaceutical Sciences, Irma Lerma Rangel College of Pharmacy, Texas A&M University, USA.
| | - Carlos F Lagos
- Chemical Biology & Drug Discovery Lab, Facultad de Medicina y Ciencia, Universidad San Sebastián, Campus Los Leones, Lota 2465 Providencia 7510157, Santiago, Chile
| | - Cristian Andrés Vilos Ortiz
- Laboratory of Nanomedicine and Targeted Delivery, Center for Medical Research, School of Medicine, Universidad de Talca, Talca, Chile
| | - Murtaza Tambuwala
- SAAD Centre for Pharmacy and Diabetes, School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine, County Londonderry, Northern Ireland BT52 1SA, UK; Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, United Kingdom.
| | | | - Flavia C Zacconi
- Departamento de Química Orgánica, Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Av. Vicuña McKenna 4860, 7820436, Macul, Santiago, Chile; Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
14
|
Bhatnagar R, Dixit NM, Yang EH, Sallam T. Cancer therapy's impact on lipid metabolism: Mechanisms and future avenues. Front Cardiovasc Med 2022; 9:925816. [PMID: 36017084 PMCID: PMC9396263 DOI: 10.3389/fcvm.2022.925816] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/22/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerotic cardiovascular disease is a growing threat among cancer patients. Not surprisingly, cancer-targeting therapies have been linked to metabolic dysregulation including changes in local and systemic lipid metabolism. Thus, tumor development and cancer therapeutics are intimately linked to cholesterol metabolism and may be a driver of increased cardiovascular morbidity and mortality in this population. Chemotherapeutic agents affect lipid metabolism through diverse mechanisms. In this review, we highlight the mechanistic and clinical evidence linking commonly used cytotoxic therapies with cholesterol metabolism and potential opportunities to limit atherosclerotic risk in this patient population. Better understanding of the link between atherosclerosis, cancer therapy, and cholesterol metabolism may inform optimal lipid therapy for cancer patients and mitigate cardiovascular disease burden.
Collapse
Affiliation(s)
- Roshni Bhatnagar
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Neal M. Dixit
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Eric H. Yang
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Tamer Sallam
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Division of Cardiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
15
|
Guha A, Stabellini N, Montero AJ. Commentary: Longitudinal changes in circulating metabolites and lipoproteins after breast cancer treatment. Front Cardiovasc Med 2022; 9:962698. [PMID: 36017091 PMCID: PMC9395698 DOI: 10.3389/fcvm.2022.962698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 07/25/2022] [Indexed: 12/03/2022] Open
Affiliation(s)
- Avirup Guha
- Cardio-Oncology Program, Department of Cardiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Nickolas Stabellini
- Graduate Education Office, Case Western Reserve University School of Medicine, Cleveland, OH, United States
- Department of Hematology-Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH, United States
- Faculdade Israelita de Ciências da Saúde Albert Einstein, Hospital Israelita Albert Einstein, São Paulo, Brazil
- Department of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Alberto J. Montero
- Department of Hematology-Oncology, University Hospitals Seidman Cancer Center, Cleveland, OH, United States
| |
Collapse
|
16
|
Mulder FVM, Peeters EFHI, Westerink J, Zwartkruis FJT, de Ranitz-Greven WL. The long-term effect of mTOR inhibition on lipid and glucose metabolism in tuberous sclerosis complex: data from the Dutch TSC registry. Orphanet J Rare Dis 2022; 17:252. [PMID: 35804402 PMCID: PMC9264703 DOI: 10.1186/s13023-022-02385-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 06/06/2022] [Indexed: 12/02/2022] Open
Abstract
Background MTOR inhibition is an effective treatment for many manifestations of tuberous sclerosis complex. Because mTOR inhibition is a disease modifying therapy, lifelong use will most likely be necessary. This study addresses the long-term effects of mTOR inhibitors on lipid and glucose metabolism and aims to provide better insight in the incidence and time course of these metabolic adverse effects in treated TSC patients.
Methods All patients who gave informed consent for the nationwide TSC Registry and were ever treated with mTOR inhibitors (sirolimus and/or everolimus) were included. Lipid profiles, HbA1c and medication were analysed in all patients before and during mTOR inhibitor treatment. Results We included 141 patients, the median age was 36 years, median use of mTOR inhibitors 5.1 years (aimed serum levels 3.0–5.0 µg/l). Total cholesterol, LDL- and HDL-cholesterol levels at baseline were similar to healthy reference data. After start of mTOR inhibition therapy, total cholesterol, LDL-cholesterol and triglycerides increased significantly and were higher compared to healthy reference population. Mean total cholesterol levels increased by 1.0 mmol/L after 3–6 months of mTOR inhibition therapy but did not increase further during follow-up. In this study, 2.5% (3/118) of patients developed diabetes (defined as an HbA1c ≥ 48 mmol/mol) during a median follow-up of 5 years. Conclusions Hypercholesterolemia is a frequent side effect of mTOR inhibition in TSC patients, and predominantly occurs within the first year of treatment. Although hyperglycemia is a frequent side effect in other indications for mTOR inhibition, incidence of diabetes mellitus in TSC patients was only 2.5%. This may reflect the difference of mTOR inhibition in patients with normal mTOR complex pathway function versus patients with overactive mTOR complex signaling due to a genetic defect (TSC patients).
Collapse
Affiliation(s)
- Femke V M Mulder
- Department of Internal Medicine, University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Evelien F H I Peeters
- Department of Internal Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jan Westerink
- Department of Internal Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Fried J T Zwartkruis
- dLAB and Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, The Netherlands
| | | |
Collapse
|
17
|
Ranallo N, Iamurri AP, Foca F, Liverani C, De Vita A, Mercatali L, Calabrese C, Spadazzi C, Fabbri C, Cavaliere D, Galassi R, Severi S, Sansovini M, Tartaglia A, Pieri F, Crudi L, Bianchini D, Barone D, Martinelli G, Frassineti GL, Ibrahim T, Calabrò L, Berardi R, Bongiovanni A. Prognostic and Predictive Role of Body Composition in Metastatic Neuroendocrine Tumor Patients Treated with Everolimus: A Real-World Data Analysis. Cancers (Basel) 2022; 14:3231. [PMID: 35805003 PMCID: PMC9264955 DOI: 10.3390/cancers14133231] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/17/2022] [Accepted: 06/23/2022] [Indexed: 02/05/2023] Open
Abstract
Neuroendocrine tumors (NETs) are rare neoplasms frequently characterized by an upregulation of the mammalian rapamycin targeting (mTOR) pathway resulting in uncontrolled cell proliferation. The mTOR pathway is also involved in skeletal muscle protein synthesis and in adipose tissue metabolism. Everolimus inhibits the mTOR pathway, resulting in blockade of cell growth and tumor progression. The aim of this study is to investigate the role of body composition indexes in patients with metastatic NETs treated with everolimus. The study population included 30 patients with well-differentiated (G1-G2), metastatic NETs treated with everolimus at the IRCCS Romagnolo Institute for the Study of Tumors (IRST) "Dino Amadori", Meldola (FC), Italy. The body composition indexes (skeletal muscle index [SMI] and adipose tissue indexes) were assessed by measuring on a computed tomography (CT) scan the cross-sectional area at L3 at baseline and at the first radiological assessment after the start of treatment. The body mass index (BMI) was assessed at baseline. The median progression-free survival (PFS) was 8.9 months (95% confidence interval [CI]: 3.4-13.7 months). The PFS stratified by tertiles was 3.2 months (95% CI: 0.9-10.1 months) in patients with low SMI (tertile 1), 14.2 months (95% CI: 2.3 months-not estimable [NE]) in patients with intermediate SMI (tertile 2), and 9.1 months (95% CI: 2.7 months-NE) in patients with high SMI (tertile 3) (p = 0.039). Similarly, the other body composition indexes also showed a statistically significant difference in the three groups on the basis of tertiles. The median PFS was 3.2 months (95% CI: 0.9-6.7 months) in underweight patients (BMI ≤ 18.49 kg/m2) and 10.1 months (95% CI: 3.7-28.4 months) in normal-weight patients (p = 0.011). There were no significant differences in terms of overall survival. The study showed a correlation between PFS and the body composition indexes in patients with NETs treated with everolimus, underlining the role of adipose and muscle tissue in these patients.
Collapse
Affiliation(s)
- Nicoletta Ranallo
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (N.R.); (L.C.)
| | - Andrea Prochoswski Iamurri
- Radiology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (A.P.I.); (D.B.)
| | - Flavia Foca
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Chiara Liverani
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (C.L.); (A.D.V.); (L.M.); (C.C.); (C.S.)
| | - Alessandro De Vita
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (C.L.); (A.D.V.); (L.M.); (C.C.); (C.S.)
| | - Laura Mercatali
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (C.L.); (A.D.V.); (L.M.); (C.C.); (C.S.)
| | - Chiara Calabrese
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (C.L.); (A.D.V.); (L.M.); (C.C.); (C.S.)
| | - Chiara Spadazzi
- Osteoncology Unit, Bioscience Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (C.L.); (A.D.V.); (L.M.); (C.C.); (C.S.)
| | - Carlo Fabbri
- Unit of Gastroenterology and Digestive Endoscopy, Forli-Cesena Hospital, AUSL Romagna, Cesena, 47121 Forli, Italy;
| | - Davide Cavaliere
- General and Oncologic Surgery Unit, Morgagni-Pierantoni Hospital, AUSL Romagna, 47121 Forlì, Italy;
| | - Riccardo Galassi
- Nuclear Medicine and Radiometabolic Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (R.G.); (S.S.); (M.S.)
| | - Stefano Severi
- Nuclear Medicine and Radiometabolic Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (R.G.); (S.S.); (M.S.)
| | - Maddalena Sansovini
- Nuclear Medicine and Radiometabolic Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (R.G.); (S.S.); (M.S.)
| | - Andreas Tartaglia
- Endocrinology Unit, Forli-Cesena Hospital, AUSL Romagna, Cesena, 47121 Forli, Italy;
| | - Federica Pieri
- Pathology Unit, Morgagni-Pierantoni Hospital, AUSL Romagna, Cesena, 47121 Forli, Italy;
| | - Laura Crudi
- Oncology Pharmacy Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori “Dino Amadori”, 47014 Meldola, Italy;
| | - David Bianchini
- Medical Physics Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, 47014 Meldola, Italy;
| | - Domenico Barone
- Radiology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (A.P.I.); (D.B.)
| | - Giovanni Martinelli
- IRCCS Istituto Romagnolo per lo Studio dei Tumori “Dino Amadori”, 47014 Meldola, Italy;
| | - Giovanni Luca Frassineti
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy;
| | - Toni Ibrahim
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Luana Calabrò
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (N.R.); (L.C.)
| | - Rossana Berardi
- Department of Medical Oncology, Università Politecnica delle Marche, AOU Ospedali Riuniti di Ancona, 60126 Ancona, Italy;
| | - Alberto Bongiovanni
- Osteoncology and Rare Tumors Center, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (N.R.); (L.C.)
| |
Collapse
|
18
|
Crocco M, Verrico A, Milanaccio C, Piccolo G, De Marco P, Gaggero G, Iurilli V, Di Profio S, Malerba F, Panciroli M, Giordano P, Calevo MG, Casalini E, Di Iorgi N, Garrè ML. Dyslipidemia in Children Treated with a BRAF Inhibitor for Low-Grade Gliomas: A New Side Effect? Cancers (Basel) 2022; 14:2693. [PMID: 35681673 PMCID: PMC9179293 DOI: 10.3390/cancers14112693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/24/2022] [Accepted: 05/27/2022] [Indexed: 02/01/2023] Open
Abstract
BRAF inhibitors, in recent years, have played a central role in the disease control of unresectable BRAF-mutated pediatric low-grade gliomas (LGGs). The aim of the study was to investigate the acute and long-term effects of vemurafenib on the lipid metabolism in children treated for an LGG. In our cohort, children treated with vemurafenib (n = 6) exhibited alterations in lipid metabolism a few weeks after starting, as was demonstrated after 1 month (n = 4) by the high plasma levels of the total cholesterol (TC = 221.5 ± 42.1 mg/dL), triglycerides (TG = 107.8 ± 44.4 mg/dL), and low-density lipoprotein (LDL = 139.5 ± 51.5 mg/dL). Despite dietary recommendations, the dyslipidemia persisted over time. The mean lipid levels of the TC (222.3 ± 34.7 mg/dL), TG (134.8 ± 83.6 mg/dL), and LDL (139.8 ± 46.9 mg/dL) were confirmed abnormal at the last follow-up (45 ± 27 months, n = 6). Vemurafenib could be associated with an increased risk of dyslipidemia. An accurate screening strategy in new clinical trials, and a multidisciplinary team, are required for the optimal management of unexpected adverse events, including dyslipidemia.
Collapse
Affiliation(s)
- Marco Crocco
- Neuroncology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (A.V.); (C.M.); (G.P.); (M.L.G.)
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, 16132 Genova, Italy; (F.M.); (M.P.); (P.G.); (E.C.); (N.D.I.)
| | - Antonio Verrico
- Neuroncology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (A.V.); (C.M.); (G.P.); (M.L.G.)
| | - Claudia Milanaccio
- Neuroncology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (A.V.); (C.M.); (G.P.); (M.L.G.)
| | - Gianluca Piccolo
- Neuroncology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (A.V.); (C.M.); (G.P.); (M.L.G.)
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, 16132 Genova, Italy; (F.M.); (M.P.); (P.G.); (E.C.); (N.D.I.)
| | - Patrizia De Marco
- UOC Genetica Medica, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy;
| | - Gabriele Gaggero
- Department of Clinical Pathology, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Valentina Iurilli
- Pharmacy Unit, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy;
| | - Sonia Di Profio
- Clinical Psychology, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy;
| | - Federica Malerba
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, 16132 Genova, Italy; (F.M.); (M.P.); (P.G.); (E.C.); (N.D.I.)
| | - Marta Panciroli
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, 16132 Genova, Italy; (F.M.); (M.P.); (P.G.); (E.C.); (N.D.I.)
| | - Paolo Giordano
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, 16132 Genova, Italy; (F.M.); (M.P.); (P.G.); (E.C.); (N.D.I.)
| | - Maria Grazia Calevo
- Epidemiology and Biostatistics Unit, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy;
| | - Emilio Casalini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, 16132 Genova, Italy; (F.M.); (M.P.); (P.G.); (E.C.); (N.D.I.)
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Natascia Di Iorgi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genova, 16132 Genova, Italy; (F.M.); (M.P.); (P.G.); (E.C.); (N.D.I.)
- Department of Pediatrics, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy
| | - Maria Luisa Garrè
- Neuroncology Unit, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy; (A.V.); (C.M.); (G.P.); (M.L.G.)
| |
Collapse
|
19
|
de Jesus M, Mohammed T, Singh M, Tiu JG, Kim AS. Etiology and Management of Dyslipidemia in Patients With Cancer. Front Cardiovasc Med 2022; 9:892335. [PMID: 35548413 PMCID: PMC9081373 DOI: 10.3389/fcvm.2022.892335] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 03/31/2022] [Indexed: 01/19/2023] Open
Abstract
Patients with cancer are now living longer than ever before due to the growth and expansion of highly effective antineoplastic therapies. Many of these patients face additional health challenges, of which cardiovascular disease (CVD) is the leading contributor to morbidity and mortality. CVD and cancer share common biological mechanisms and risk factors, including lipid abnormalities. A better understanding of the relationship between lipid metabolism and cancer can reveal strategies for cancer prevention and CVD risk reduction. Several anticancer treatments adversely affect lipid levels, increasing triglycerides and/or LDL-cholesterol. The traditional CVD risk assessment tools do not include cancer-specific parameters and may underestimate the true long-term CVD risk in this patient population. Statins are the mainstay of therapy in both primary and secondary CVD prevention. The role of non-statin therapies, including ezetimibe, PCSK9 inhibitors, bempedoic acid and icosapent ethyl in the management of lipid disorders in patients with cancer remains largely unknown. A contemporary cancer patient needs a personalized comprehensive cardiovascular assessment, management of lipid abnormalities, and prevention of late CVD to achieve optimal overall outcomes.
Collapse
Affiliation(s)
- Mikhail de Jesus
- Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Turab Mohammed
- Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Meghana Singh
- Department of Medicine, University of Connecticut School of Medicine, Farmington, CT, United States
| | - John G. Tiu
- Department of Medicine, Calhoun Cardiology Center, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Agnes S. Kim
- Department of Medicine, Calhoun Cardiology Center, University of Connecticut School of Medicine, Farmington, CT, United States
| |
Collapse
|
20
|
Joharatnam-Hogan N, Chambers P, Dhatariya K, Board R. A guideline for the outpatient management of glycaemic control in people with cancer. Diabet Med 2022; 39:e14636. [PMID: 34240470 DOI: 10.1111/dme.14636] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 12/17/2022]
Abstract
Individuals with cancer are at increased risk of developing new-onset diabetes mellitus and hyperglycaemia, and an estimated 20% of people with cancer already have an underlying diagnosis of diabetes mellitus. People with both cancer and diabetes may have an increased risk of toxicities, hospital admissions and morbidity, with hyperglycaemia potentially attenuating the efficacy of chemotherapy often secondary to dose reductions and early cessation. Numerous studies have demonstrated that hyperglycaemia is prognostic of worse overall survival and risk of cancer recurrence. These guidelines aim to provide the oncology/haemato-oncology and diabetes multidisciplinary teams with the tools to manage people with diabetes commencing anti-cancer/glucocorticoid therapy, as well as identifying individuals without a known diagnosis of diabetes who are at risk of developing hyperglycaemia and new-onset diabetes.
Collapse
Affiliation(s)
| | - Pinkie Chambers
- University College London Hospital NHS Foundation Trust, London, UK
| | - Ketan Dhatariya
- Norfolk and Norwich Hospitals NHS Foundation Trust, London, UK
| | - Ruth Board
- Lancashire Teaching Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
21
|
A Population-Based Study of Cardiovascular Disease Mortality in Italian Cancer Patients. Cancers (Basel) 2021; 13:cancers13235903. [PMID: 34885016 PMCID: PMC8656614 DOI: 10.3390/cancers13235903] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 11/18/2021] [Accepted: 11/22/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Research suggests that lengthening the prognosis in cancer patients (which is certainly a positive thing) requires greater attention to be paid to the onset of other pathologies. The increase in cardiovascular mortality in cancer patients (due to both exposure to risk factors and the side effects of cytotoxic drugs) requires greater collaboration between oncologists and cardiologists, and the integration of shared follow-up paths. Particular attention to the follow-up of cancer patients can reduce this risk: in the present study, patients presented an excessive risk of cardiovascular mortality only in the first two years from cancer diagnosis. Abstract The present research describes 25 years of cardiovascular mortality in a cohort of patients in Northern Italy. The study included patients with malignant cancer enrolled in the period of 1996–2019, and describes cardiovascular and cancer mortality in relation to sex, age, year of diagnosis, months of survivorship, tumor site, and standardized mortality ratio (SMR). Out of 67,173 patients, 38,272 deaths (57.7%) were recorded: 4466 from cardiovascular disease (CVD) (6.6%), and 28,579 (42.6%) from cancer. The proportion of CVD death increased from 4.5% in the first two years after diagnosis, to 7.3% after more than 10 years, while the proportion of deaths from cancer decreased from 70.5% to 9.4%. The CVD SMR comparing cancer patients with the general population was 0.87 (95% CI: 0.82–0.92) in 1996–1999, rising to 0.95 (95% CI: 0.84–1.08) in 2015–2019, without differences in terms of sex or age. The risk of dying from CVD was higher compared with the general population (SMR 1.31; 95% CI: 1.24–1.39) only in the first two years after diagnosis. The trend over time underscored that CVD deaths increased in patients with breast, bladder, prostate, and colorectal cancers, and, in the more recent period, for kidney cancer and melanoma patients. Our data confirmed that cardiovascular mortality is an important issue in the modern management of cancer patients, suggesting the need for an extensive interdisciplinary approach.
Collapse
|
22
|
Clark H, Schulte R, Haggstrom AN. Severe hypertriglyceridemia following sirolimus use in an infant. Pediatr Dermatol 2021; 38:1581-1582. [PMID: 34749436 DOI: 10.1111/pde.14853] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Inhibitors of mammalian target of rapamycin function to downregulate cell growth and proliferation and have off-label use in pediatrics for vascular malformations. Hypertriglyceridemia is a known side effect of mammalian target of rapamycin (mTOR) inhibitors. Further studies to better understand the incidence and treatment of hypertriglyceridemia in infants and neonates are warranted.
Collapse
Affiliation(s)
- Hannah Clark
- Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Rachael Schulte
- Departments of Pediatrics and Dermatology, Indiana University, Indianapolis, Indiana, USA
| | - Anita N Haggstrom
- Division of Pediatric Hematology/Oncology, Indiana University, Indianapolis, Indiana, USA
| |
Collapse
|
23
|
Durrani IA, Bhatti A, John P. The prognostic outcome of 'type 2 diabetes mellitus and breast cancer' association pivots on hypoxia-hyperglycemia axis. Cancer Cell Int 2021; 21:351. [PMID: 34225729 PMCID: PMC8259382 DOI: 10.1186/s12935-021-02040-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
Type 2 diabetes mellitus and breast cancer are complex, chronic, heterogeneous, and multi-factorial diseases; with common risk factors including but not limited to diet, obesity, and age. They also share mutually inclusive phenotypic features such as the metabolic deregulations resulting from hyperglycemia, hypoxic conditions and hormonal imbalances. Although, the association between diabetes and cancer has long been speculated; however, the exact molecular nature of this link remains to be fully elucidated. Both the diseases are leading causes of death worldwide and a causal relationship between the two if not addressed, may translate into a major global health concern. Previous studies have hypothesized hyperglycemia, hyperinsulinemia, hormonal imbalances and chronic inflammation, as some of the possible grounds for explaining how diabetes may lead to cancer initiation, yet further research still needs to be done to validate these proposed mechanisms. At the crux of this dilemma, hyperglycemia and hypoxia are two intimately related states involving an intricate level of crosstalk and hypoxia inducible factor 1, at the center of this, plays a key role in mediating an aggressive disease state, particularly in solid tumors such as breast cancer. Subsequently, elucidating the role of HIF1 in establishing the diabetes-breast cancer link on hypoxia-hyperglycemia axis may not only provide an insight into the molecular mechanisms underlying the association but also, illuminate on the prognostic outcome of the therapeutic targeting of HIF1 signaling in diabetic patients with breast cancer or vice versa. Hence, this review highlights the critical role of HIF1 signaling in patients with both T2DM and breast cancer, potentiates its significance as a prognostic marker in comorbid patients, and further discusses the potential prognostic outcome of targeting HIF1, subsequently establishing the pressing need for HIF1 molecular profiling-based patient selection leading to more effective therapeutic strategies emerging from personalized medicine.
Collapse
Affiliation(s)
- Ilhaam Ayaz Durrani
- Atta-ur-Rehman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| | - Attya Bhatti
- Atta-ur-Rehman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan.
| | - Peter John
- Atta-ur-Rehman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), H-12, Islamabad, Pakistan
| |
Collapse
|
24
|
Rashid MM, Lee H, Park J, Jung BH. Comparative metabolomics and lipidomics study to evaluate the metabolic differences between first- and second-generation mammalian or mechanistic target of rapamycin inhibitors. Biomed Chromatogr 2021; 35:e5190. [PMID: 34101862 DOI: 10.1002/bmc.5190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/30/2021] [Accepted: 06/04/2021] [Indexed: 12/29/2022]
Abstract
Mammalian or mechanistic target of rapamycin (mTOR) drives its fundamental cellular functions through two distinct catalytic subunits, mTORC1 and mTORC2, and is frequently dysregulated in most cancers. To treat cancers, developed mTOR inhibitors have been classified into first and second generations based on their ability to inhibit single (first-generation) and dual (second-generation) mTOR subunits. However, the underlying metabolic differences due to the effects of first- and second-generation mTOR inhibitors have not been clearly evaluated. In this study, rapamycin (sirolimus) and AZD8055 and PP242 were selected as first- and second-generation mTOR inhibitors, respectively, to evaluate the metabolic differences due to these two generations of mTOR inhibitors after a single oral dose using untargeted metabolomics and lipidomics approaches. The metabolic differences at each time point were compared using multivariate analysis. The multivariate and data analyses showed that metabolic disparity was more prominent within 8 h after drug administration and a broad class of metabolites were affected by the administration of both generations of mTOR inhibitors. Among the metabolite classes, changes in the pattern of fatty acids and glycerophospholipids were opposite, specifically at 4 and 8 h between the two generations of mTOR inhibitors. We speculate that the inhibition of the mTORC2 subunit by the second-generation mTOR inhibitor may have resulted in a distinct metabolic pattern between the first- and second-generation inhibitors. Finally, the findings of this study could assist in a more detailed understanding of the key metabolic differences caused by first- and second-generation mTOR inhibitors.
Collapse
Affiliation(s)
- Md Mamunur Rashid
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, South Korea.,Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, South Korea
| | - Hyunbeom Lee
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, South Korea
| | - Jinyoung Park
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, South Korea
| | - Byung Hwa Jung
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, South Korea.,Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, South Korea
| |
Collapse
|
25
|
Endocrine Toxicities of Antineoplastic Therapy. Cancers (Basel) 2021; 13:cancers13020294. [PMID: 33466843 PMCID: PMC7830439 DOI: 10.3390/cancers13020294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 01/12/2021] [Indexed: 11/16/2022] Open
Abstract
In recent years, the prognosis of many solid tumors has improved markedly thanks to new treatment strategies, including tyrosine kinase inhibitors (TKIs) and immunotherapy [...].
Collapse
|
26
|
Silvestris N, Argentiero A, Beretta GD, Di Bartolo P, Montagnani M, Danesi R, Ferrari P, D'Oronzo S, Gori S, Russo A, Acquati S, Gallo M. Management of metabolic adverse events of targeted therapies and immune checkpoint inhibitors in cancer patients: an Associazione Italiana Oncologia Medica (AIOM)/Associazione Medici Diabetologi (AMD)/Società Italiana Farmacologia (SIF) multidisciplinary consensus position paper. Crit Rev Oncol Hematol 2020; 154:103066. [PMID: 32853883 DOI: 10.1016/j.critrevonc.2020.103066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/16/2022] Open
Abstract
The growing insights in the next-generation immunotherapy and the state-of-the-art advancement in targeted-agents significantly improved clinical outcome of cancer patients by pointing towards a unexplored Achilles' heel. Novel toxicity profiles have been uncovered, representing unmet medical needs. Thus, a panel of expert provide comprehensive pharmacological and clinical evidence, to provide a patient-tailored approach to metabolic adverse events associated with novel anti-cancer treatments. Prompted by the need of a multidisciplinary cooperation, a working group of Associazione Italiana Oncologia Medica (AIOM), Associazione Medici Diabetologi (AMD) and Società Italiana Farmacologia (SIF) examined the available literature data. The identification of patient risk profile and the characterization of metabolic effects of novel anti-tumour drugs is clearly a clinical challenge that can be addressed by a multidisciplinary clinical approach. Therefore, this review pinpoints the relevance of the challenging profiling of the patient suffering from dysmetabolic conditions induced by the novel therapeutics in medical oncology.
Collapse
Affiliation(s)
- Nicola Silvestris
- IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, Italy; Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", Bari, Italy.
| | | | | | - Paolo Di Bartolo
- Diabetology Unit, Rete Clinica di Diabetologia Aziendale - Dipartimento, Internistico di Ravenna - AUSL Romagna, Ravenna, Italy
| | - Monica Montagnani
- Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | - Pietro Ferrari
- Palliative Care Unit, Istituti Clinici Scientifici Maugeri SPA SB, IRCCS (PV), Italy
| | - Stella D'Oronzo
- IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, Italy; Department of Biomedical Sciences and Human Oncology, University of Bari "Aldo Moro", Bari, Italy
| | - Stefania Gori
- Oncologia Medica, IRCCS Ospedale Don Calabria-Sacro Cuore di Negrar, Verona, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Italy
| | - Silvia Acquati
- Endocrinology Unit, Ospedale Pierantoni-Morgagni, Forlì, Italy
| | - Marco Gallo
- Oncological Endocrinology Unit, Department of Medical Sciences, University of Torino, AOU Città della Salute e della Scienza di Torino, Torino, Italy
| |
Collapse
|
27
|
Srivastava SP, Goodwin JE. Cancer Biology and Prevention in Diabetes. Cells 2020; 9:cells9061380. [PMID: 32498358 PMCID: PMC7349292 DOI: 10.3390/cells9061380] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/25/2020] [Accepted: 05/30/2020] [Indexed: 02/07/2023] Open
Abstract
The available evidence suggests a complex relationship between diabetes and cancer. Epidemiological data suggest a positive correlation, however, in certain types of cancer, a more complex picture emerges, such as in some site-specific cancers being specific to type I diabetes but not to type II diabetes. Reports share common and differential mechanisms which affect the relationship between diabetes and cancer. We discuss the use of antidiabetic drugs in a wide range of cancer therapy and cancer therapeutics in the development of hyperglycemia, especially antineoplastic drugs which often induce hyperglycemia by targeting insulin/IGF-1 signaling. Similarly, dipeptidyl peptidase 4 (DPP-4), a well-known target in type II diabetes mellitus, has differential effects on cancer types. Past studies suggest a protective role of DPP-4 inhibitors, but recent studies show that DPP-4 inhibition induces cancer metastasis. Moreover, molecular pathological mechanisms of cancer in diabetes are currently largely unclear. The cancer-causing mechanisms in diabetes have been shown to be complex, including excessive ROS-formation, destruction of essential biomolecules, chronic inflammation, and impaired healing phenomena, collectively leading to carcinogenesis in diabetic conditions. Diabetes-associated epithelial-to-mesenchymal transition (EMT) and endothelial-to-mesenchymal transition (EndMT) contribute to cancer-associated fibroblast (CAF) formation in tumors, allowing the epithelium and endothelium to enable tumor cell extravasation. In this review, we discuss the risk of cancer associated with anti-diabetic therapies, including DPP-4 inhibitors and SGLT2 inhibitors, and the role of catechol-o-methyltransferase (COMT), AMPK, and cell-specific glucocorticoid receptors in cancer biology. We explore possible mechanistic links between diabetes and cancer biology and discuss new therapeutic approaches.
Collapse
Affiliation(s)
- Swayam Prakash Srivastava
- Department of Pediatrics, Yale University School of Medicine, Yale University, New Haven, CT 06520-8064, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520-8066, USA
- Correspondence: (S.P.S.); (J.E.G.)
| | - Julie E. Goodwin
- Department of Pediatrics, Yale University School of Medicine, Yale University, New Haven, CT 06520-8064, USA
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT 06520-8066, USA
- Correspondence: (S.P.S.); (J.E.G.)
| |
Collapse
|
28
|
Tricistronic expression of MOAP-1, Bax and RASSF1A in cancer cells enhances chemo-sensitization that requires BH3L domain of MOAP-1. J Cancer Res Clin Oncol 2020; 146:1751-1764. [DOI: 10.1007/s00432-020-03231-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 04/21/2020] [Indexed: 01/15/2023]
|
29
|
Zhang J, Deng Y, Khoo BL. Fasting to enhance Cancer treatment in models: the next steps. J Biomed Sci 2020; 27:58. [PMID: 32370764 PMCID: PMC7201989 DOI: 10.1186/s12929-020-00651-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 04/22/2020] [Indexed: 02/06/2023] Open
Abstract
Short-term fasting (STF) is a technique to reduce nutrient intake for a specific period. Since metabolism plays a pivotal role in tumor progression, it can be hypothesized that STF can improve the efficacy of chemotherapy. Recent studies have demonstrated the efficacy of STF in cell and animal tumor models. However, large-scale clinical trials must be conducted to verify the safety and effectiveness of these diets. In this review, we re-examine the concept of how metabolism affects pathophysiological pathways. Next, we provided a comprehensive discussion of the specific mechanisms of STF on tumor progression, derived through studies carried out with tumor models. There are currently at least four active clinical trials on fasting and cancer treatment. Based on these studies, we highlight the potential caveats of fasting in clinical applications, including the onset of metabolic syndrome and other metabolic complications during chemotherapy, with a particular focus on the regulation of the epithelial to mesenchymal pathway and cancer heterogeneity. We further discuss the advantages and disadvantages of the current state-of-art tumor models for assessing the impact of STF on cancer treatment. Finally, we explored upcoming fasting strategies that could complement existing chemotherapy and immunotherapy strategies to enable personalized medicine. Overall, these studies have the potential for breakthroughs in cancer management.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong
| | - Yanlin Deng
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong
| | - Bee Luan Khoo
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon Tong, Hong Kong.
| |
Collapse
|
30
|
Lai S, Amabile MI, Mazzaferro S, Mitterhofer AP, Mazzarella A, Galani A, Imbimbo G, Cianci R, Pasquali M, Molfino A. Effects of sunitinib on endothelial dysfunction, metabolic changes, and cardiovascular risk indices in renal cell carcinoma. Cancer Med 2020; 9:3752-3757. [PMID: 32270594 PMCID: PMC7286450 DOI: 10.1002/cam4.2910] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/31/2019] [Indexed: 12/19/2022] Open
Abstract
Background Sunitinib is a standard treatment for metastatic renal cell carcinoma (RCC). Currently, the data available on the effects of sunitinib on endothelial dysfunction, metabolic changes, and cardiovascular (CV) risk factors are limited, and we aimed to evaluate these aspects in patients with RCC after a short period of treatment. Methods Patients affected by metastatic RCC were enrolled and evaluated before starting sunitinib (T0) and after 40 days of treatment (T1) by the flow‐mediated dilation (FMD), carotid intima media thickness (IMT), ankle‐brachial pressure index (ABI), and 24‐hour proteinuria. We also assessed serum metabolic and nutritional parameters at T0 and T1. Results Twenty patients (7 female), with a mean age of 61.4 ± 12.0 years, were studied. Overtime, we observed a reduction in estimated glomerular filtration rate (P = .002), FMD (P = .001) and in fasting plasma glucose levels (P = .04), as well as an increase in plasma insulin (P < .001), HOMA‐IR (P < .01), and serum total cholesterol levels (P = .01). Moreover at T1 we found a significant increase in systolic and diastolic blood pressure (P ≤ .001) and 24‐hour proteinuria (P < .001) compared to baseline, whereas no changes in IMT and ABI were detected. Conclusion The changes observed overtime after sunitinib treatment in terms of markers of early endothelial dysfunction, blood pressure, as well as in glucose/insulin metabolism and proteinuria may contribute to increase CV risk in RCC patients and suggest a strict follow‐up in this setting. Larger evidences are mandatory to confirm our observations.
Collapse
Affiliation(s)
- Silvia Lai
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Maria Ida Amabile
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Sandro Mazzaferro
- Department of Cardiovascular, Respiratory, Nephrological, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, Rome, Italy
| | - Anna Paola Mitterhofer
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Angelo Mazzarella
- Department of Cardiovascular, Respiratory, Nephrological, Anesthesiology and Geriatric Sciences, Sapienza University of Rome, Rome, Italy
| | - Alessandro Galani
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Giovanni Imbimbo
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Rosario Cianci
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Marzia Pasquali
- Nephrology and Dialysis Unit, Policlinico Umberto I, Rome, Italy
| | - Alessio Molfino
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | | |
Collapse
|
31
|
Zafar MI. Suitability of APINCH high-risk medications use in diabetes mellitus. Eur J Pharmacol 2020; 867:172845. [DOI: 10.1016/j.ejphar.2019.172845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/15/2019] [Accepted: 12/03/2019] [Indexed: 12/17/2022]
|
32
|
Duvillié B, Kourdoughli R, Druillennec S, Eychène A, Pouponnot C. Interplay Between Diabetes and Pancreatic Ductal Adenocarcinoma and Insulinoma: The Role of Aging, Genetic Factors, and Obesity. Front Endocrinol (Lausanne) 2020; 11:563267. [PMID: 33101198 PMCID: PMC7556217 DOI: 10.3389/fendo.2020.563267] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/14/2020] [Indexed: 12/11/2022] Open
Abstract
Epidemiologic analyses have shed light on an association between type 2 diabetes (T2D) and pancreatic ductal adenocarcinoma (PDAC). Recent data also suggest a potential relationship between T2D and insulinoma. Under rare circumstances, type 1 diabetes (T1D) can also be implicated in tumorigenesis. The biological mechanisms underlying such relationships are extremely complex. Some genetic factors contributing to the development of T2D are shared with pancreatic exocrine and endocrine tumors. Obesity and overweight can also contribute to the initiation and severity of T2D, while aging may influence both endocrine and exocrine tumors. Finally, pharmacological treatments of T2D may have an impact on PDAC. On the other hand, some treatments for insulinoma can trigger diabetes. In the present minireview, we discuss the cellular and molecular mechanisms that could explain these interactions. This analysis may help to define new potential therapeutic strategies.
Collapse
Affiliation(s)
- Bertrand Duvillié
- Department of Signaling, Radiobiology and Cancer, Institut Curie, Orsay, France
- INSERM U1021, Centre Universitaire, Orsay, France
- CNRS UMR 3347, Centre Universitaire, Orsay, France
- Université Paris-Saclay, Orsay, France
- PSL Research University, Paris, France
- *Correspondence: Bertrand Duvillié,
| | - Rayane Kourdoughli
- Department of Signaling, Radiobiology and Cancer, Institut Curie, Orsay, France
- INSERM U1021, Centre Universitaire, Orsay, France
- CNRS UMR 3347, Centre Universitaire, Orsay, France
- Université Paris-Saclay, Orsay, France
- PSL Research University, Paris, France
| | - Sabine Druillennec
- Department of Signaling, Radiobiology and Cancer, Institut Curie, Orsay, France
- INSERM U1021, Centre Universitaire, Orsay, France
- CNRS UMR 3347, Centre Universitaire, Orsay, France
- Université Paris-Saclay, Orsay, France
- PSL Research University, Paris, France
| | - Alain Eychène
- Department of Signaling, Radiobiology and Cancer, Institut Curie, Orsay, France
- INSERM U1021, Centre Universitaire, Orsay, France
- CNRS UMR 3347, Centre Universitaire, Orsay, France
- Université Paris-Saclay, Orsay, France
- PSL Research University, Paris, France
| | - Celio Pouponnot
- Department of Signaling, Radiobiology and Cancer, Institut Curie, Orsay, France
- INSERM U1021, Centre Universitaire, Orsay, France
- CNRS UMR 3347, Centre Universitaire, Orsay, France
- Université Paris-Saclay, Orsay, France
- PSL Research University, Paris, France
| |
Collapse
|
33
|
Goldberg RB, Chait A. A Comprehensive Update on the Chylomicronemia Syndrome. Front Endocrinol (Lausanne) 2020; 11:593931. [PMID: 33193106 PMCID: PMC7644836 DOI: 10.3389/fendo.2020.593931] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 10/02/2020] [Indexed: 12/13/2022] Open
Abstract
The chylomicronemia syndrome is characterized by severe hypertriglyceridemia and fasting chylomicronemia and predisposes affected individuals to acute pancreatitis. When due to very rare monogenic mutations in the genes encoding the enzyme, lipoprotein lipase, or its regulators, APOC2, APOA5, GPIHBP1, and LMF1, it is referred to as the familial chylomicronemia syndrome. Much more frequently, the chylomicronemia syndrome results from a cluster of minor genetic variants causing polygenic hypertriglyceridemia, which is exacerbated by conditions or medications which increase triglyceride levels beyond the saturation point of triglyceride removal systems. This situation is termed the multifactorial chylomicronemia syndrome. These aggravating factors include common conditions such as uncontrolled diabetes, overweight and obesity, alcohol excess, chronic kidney disease and pregnancy and several medications, including diuretics, non-selective beta blockers, estrogenic compounds, corticosteroids, protease inhibitors, immunosuppressives, antipsychotics, antidepressants, retinoids, L-asparaginase, and propofol. A third uncommon cause of the chylomicronemia syndrome is familial forms of partial lipodystrophy. Development of pancreatitis is the most feared complication of the chylomicronemia syndrome, but the risk of cardiovascular disease as well as non-alcoholic steatohepatitis is also increased. Treatment consists of dietary fat restriction and weight reduction combined with the use of triglyceride lowering medications such as fibrates, omega 3 fatty acids and niacin. Effective management of aggravating factors such as improving diabetes control, discontinuing alcohol and replacing or reducing the dose of medications that raise triglyceride levels is essential. Importantly, many if not most cases of the chylomicronemia syndrome can be prevented by effective identification of polygenic hypertriglyceridemia in people with conditions that increase its likelihood or before starting medications that may increase triglyceride levels. Several new pharmacotherapeutic agents are being tested that are likely to considerably improve treatment of hypertriglyceridemia in people at risk.
Collapse
Affiliation(s)
- Ronald B. Goldberg
- Departments of Medicine, Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, United States
- *Correspondence: Ronald B. Goldberg,
| | - Alan Chait
- Department of Medicine, University of Washington, Seattle, WA, United States
| |
Collapse
|
34
|
Chiu WL, Churilov L, Lim CH, Tan A, Nedumannil R, Lau LH, Lew J, Hachem M, Kong A, Robbins R, Sutcliffe H, Lam Q, Lee A, Djukiadmodjo F, Nanayakkara N, Zajac JD, Ekinci EI. Routine HbA1c among hematology and oncology inpatients: Diabetes-status and hospital-outcomes. Diabetes Res Clin Pract 2019; 152:71-78. [PMID: 31082446 DOI: 10.1016/j.diabres.2019.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/25/2019] [Accepted: 05/03/2019] [Indexed: 01/08/2023]
Abstract
AIMS Using routine HbA1c measurement to determine the prevalence of diabetes mellitus (known and previously unrecognized) and their hospital outcomes among hematology and oncology inpatients. METHODS This was a prospective, observational study. Routine automated HbA1c testing was performed in all hematology and oncology inpatients aged ≥54 years at a tertiary hospital, July 2013-January 2015. The outcome measures were: (i) prevalence of known and previously unrecognized diabetes, and (ii) hospital outcomes: length-of-stay (LOS), intensive-care-unit (ICU) admission, 30-day/18-month readmission, and 18-month mortality. RESULTS Over the 18-month study period, 1076 inpatients aged ≥54 years were admitted to hematology (n = 298) and oncology (n = 778) units: 21% had known diabetes and 7% had previously unrecognized diabetes. Patients with known diabetes had a longer LOS (IRR: 1.18, 95%CI: 1.02-1.37, p = 0.03), compared to those without diabetes, adjusting for age, hemoglobin level, estimated-glomerular-filtration-rate, admission specialty unit, Charlson's comorbidity index score, and glucocorticoid exposure. No significant differences were observed in ICU admission, 30-day/18-month readmission, and 18-month mortality among patients with known, previously unrecognized and no diabetes (p ≥ 0.05). CONCLUSIONS Approximately one in five hematology or oncology inpatients aged ≥54 years had known diabetes, and one in fourteen had previously unrecognized diabetes. Those with known diabetes had a longer hospital stay. Routine HbA1c measurement is can be useful for identifying previously unrecognized diabetes, particularly among patients with high glucocorticoid exposure. Further study is required to determine cost-effectiveness in screening for unrecognized diabetes and optimal management of these patients.
Collapse
Affiliation(s)
- Wei-Ling Chiu
- Department of Endocrinology, Austin Health, 145 Studley Road, Heidelberg, Victoria 3084, Australia.
| | - Leonid Churilov
- The Florey Institute of Neuroscience & Mental Health, Heidelberg, Victoria 3084, Australia; University of Melbourne - Austin Health, Department of Medicine, Heidelberg, Victoria 3084, Australia.
| | - Chee-Hau Lim
- Department of Endocrinology, Austin Health, 145 Studley Road, Heidelberg, Victoria 3084, Australia
| | - Alanna Tan
- Department of Endocrinology, Austin Health, 145 Studley Road, Heidelberg, Victoria 3084, Australia.
| | - Rithin Nedumannil
- Department of Endocrinology, Austin Health, 145 Studley Road, Heidelberg, Victoria 3084, Australia
| | - Lik-Hui Lau
- Department of Endocrinology, Austin Health, 145 Studley Road, Heidelberg, Victoria 3084, Australia
| | - Jeremy Lew
- University of Melbourne, Grattan Street, Parkville, Victoria 3010, Australia
| | - Mariam Hachem
- University of Melbourne - Austin Health, Department of Medicine, Heidelberg, Victoria 3084, Australia.
| | - Alvin Kong
- University of Melbourne, Grattan Street, Parkville, Victoria 3010, Australia
| | - Raymond Robbins
- Department of Strategy, Quality & Service Redesign, Austin Health, 145 Studley Road, Heidelberg, Victoria 3084, Australia.
| | - Harvey Sutcliffe
- Pathology IT Service, Austin Pathology, Heidelberg, Victoria 3084, Australia.
| | - Que Lam
- Pathology IT Service, Austin Pathology, Heidelberg, Victoria 3084, Australia.
| | - Andrew Lee
- Clinical Informatics Unit, Austin Health, Heidelberg, Victoria 3084, Australia.
| | - Frida Djukiadmodjo
- Department of Endocrinology, Austin Health, 145 Studley Road, Heidelberg, Victoria 3084, Australia
| | - Natalie Nanayakkara
- Department of Endocrinology, Austin Health, 145 Studley Road, Heidelberg, Victoria 3084, Australia.
| | - Jeffrey D Zajac
- Department of Endocrinology, Austin Health, 145 Studley Road, Heidelberg, Victoria 3084, Australia; University of Melbourne - Austin Health, Department of Medicine, Heidelberg, Victoria 3084, Australia.
| | - Elif I Ekinci
- Department of Endocrinology, Austin Health, 145 Studley Road, Heidelberg, Victoria 3084, Australia; University of Melbourne - Austin Health, Department of Medicine, Heidelberg, Victoria 3084, Australia.
| |
Collapse
|
35
|
Abstract
We report a case of severe hypertriglyceridemia associated with an everolimus drug-eluting stent in an infant with pulmonary vein stenosis. We review from current literature the mechanisms by which everolimus may cause dyslipidaemia, pharmacokinetics of everolimus in drug-eluting stents, and treatments of hypertriglyceridemia. This case demonstrates the need to closely monitor serum triglyceride levels after everolimus drug-eluting stent placement in infants.
Collapse
|
36
|
Vernieri C, Pusceddu S, Fucà G, Indelicato P, Centonze G, Castagnoli L, Ferrari E, Ajazi A, Pupa S, Casola S, Foiani M, Mazzaferro V, Pruneri G, Milione M, de Braud F. Impact of systemic and tumor lipid metabolism on everolimus efficacy in advanced pancreatic neuroendocrine tumors (pNETs). Int J Cancer 2019; 144:1704-1712. [PMID: 30520016 DOI: 10.1002/ijc.32042] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 10/26/2018] [Accepted: 11/19/2018] [Indexed: 11/06/2022]
Abstract
The mTOR inhibitor everolimus is effective against advanced pancreatic neuroendocrine tumors (pNETs). However, it can cause metabolic adverse events, such as hyperglycemia, hypertriglyceridemia and hypercholesterolemia. In this work we aimed at evaluating the impact of systemic and tumor lipid metabolism on everolimus efficacy. We carried out a monocentric, retrospective study to correlate plasma triglyceride and cholesterol levels with the progression free survival (PFS) of advanced pNET patients treated with everolimus. In formalin fixed, paraffin embedded (FFPE) tumor specimens, we also assessed by mRNA quantification and immunohistochemistry the expression of acetyl-CoA carboxylase 1 (ACC1) and fatty acid synthase (FASN), two enzymes crucially involved in fatty acid biosynthesis, and we analyzed their impact on PFS. We evaluated 58 consecutive pNET patients who started everolimus between December 2006 and January 2015. Patients with higher plasma triglycerides during the first 3 months of treatment had an increased risk of disease progression (aHR 3.08, 95% CIs 1.15-8.21; p = 0.025). In 23 FFPE tumor specimens amenable for IHC evaluations, we found a positive correlation between ACC1 and FASN at both mRNA (r = 0.87, p = 0.00045) and protein (r = 0.68, p = 0.0004) level. Patients with higher ACC1 protein expression in metastatic lesions had significantly lower PFS when compared to patients with lower ACC1 levels (5.5 vs. 36 months; aHR 4.49, 95% CIs 1.08-18.72; p = 0.039). In conclusion, systemic and tumor lipid metabolism are associated with the PFS of everolimus-treated patients with advanced pNETs; based on these findings, dietary and pharmacological interventions targeting lipid metabolism could improve everolimus efficacy in this patient population.
Collapse
Affiliation(s)
- Claudio Vernieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy.,IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Sara Pusceddu
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy
| | - Giovanni Fucà
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy
| | - Pietro Indelicato
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy
| | - Giovanni Centonze
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy
| | - Lorenzo Castagnoli
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy
| | - Elisa Ferrari
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Arta Ajazi
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Serenella Pupa
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy
| | - Stefano Casola
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Marco Foiani
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy.,Oncology and Haemato-Oncology Department, University of Milan, Milan, Italy
| | - Vincenzo Mazzaferro
- Hepato-Bilio-Pancreatic Surgery and Liver Transplantation, Fondazione IRCCS Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy
| | - Giancarlo Pruneri
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy.,School of Medicine, University of Milan, Milan, Italy
| | - Massimo Milione
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy.,Oncology and Haemato-Oncology Department, University of Milan, Milan, Italy
| |
Collapse
|
37
|
Santos AP, Santos AC, Castro C, Raposo L, Pereira SS, Torres I, Henrique R, Cardoso H, Monteiro MP. Visceral Obesity and Metabolic Syndrome Are Associated with Well-Differentiated Gastroenteropancreatic Neuroendocrine Tumors. Cancers (Basel) 2018; 10:cancers10090293. [PMID: 30150555 PMCID: PMC6162651 DOI: 10.3390/cancers10090293] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 08/21/2018] [Accepted: 08/24/2018] [Indexed: 02/06/2023] Open
Abstract
The determinants for gastroenteropancreatic neuroendocrine tumors (GEP-NET) recent burden are matters of debate. Obesity and metabolic syndrome (MetS) are well established risks for several cancers even though no link with GEP-NETs was yet established. Our aim in this study was to investigate whether well-differentiated GEP-NETs were associated with obesity and MetS. Patients with well-differentiated GEP-NETs (n = 96) were cross-matched for age, gender, and district of residence with a control group (n = 96) derived from the general population in a case-control study. Patients presented gastro-intestinal (75.0%) or pancreatic (22.9%) tumors, grade G1 (66.7%) or G2 (27.1%) with localized disease (31.3%), regional metastasis (16.7%) or distant metastasis (43.8%) at diagnosis, and 45.8% had clinical hormonal syndromes. MetS was defined according to Joint Interim Statement (JIS) criteria. Well-differentiated GEP-NETs were associated with MetS criteria as well as the individual components' waist circumference, fasting triglycerides, and fasting plasma glucose (p = 0.003, p = 0.002, p = 0.011 and p < 0.001, respectively). The likelihood of the association was higher when the number of individual MetS components was greater than four. MetS and some individual MetS components including visceral obesity, dyslipidemia, and increased fasting glucose are associated with well-differentiated GEP-NET. This data provides a novel insight in unraveling the mechanisms leading to GEP-NET disease.
Collapse
Affiliation(s)
- Ana P Santos
- Department of Endocrinology of Portuguese Oncology Institute of Porto (IPO-Porto) & Clinical Research Unit-Research Center of IPO-Porto, 4200-072 Porto, Portugal.
| | - Ana C Santos
- Department of Public Health and Forensic Sciences and Medical Education, Unit of Clinical Epidemiology, Predictive Medicine and Public Health, University of Porto Medical School, 4200-319 Porto, Portugal.
- EPIUnit-Instituto de Saúde Pública, Universidade do Porto, 4050-600 Porto, Portugal.
| | - Clara Castro
- EPIUnit-Instituto de Saúde Pública, Universidade do Porto, 4050-600 Porto, Portugal.
- Department of Epidemiology of Portuguese Oncology Institute of Porto (IPO-Porto), 4200-072 Porto, Portugal.
| | - Luís Raposo
- EPIUnit-Instituto de Saúde Pública, Universidade do Porto, 4050-600 Porto, Portugal.
| | - Sofia S Pereira
- Endocrine, Cardiovascular & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal.
- Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal.
| | - Isabel Torres
- Department of Endocrinology of Portuguese Oncology Institute of Porto (IPO-Porto) & Clinical Research Unit-Research Center of IPO-Porto, 4200-072 Porto, Portugal.
| | - Rui Henrique
- Department of Pathology of Portuguese Oncology Institute of Porto (IPO-Porto) & Cancer Biology and Epigenetics Group-Research Center of IPO-Porto, 4200-072 Porto, Portugal.
- Department of Pathology and Molecular Immunology of Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal.
| | - Helena Cardoso
- Endocrine, Cardiovascular & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal.
- Department of Endocrinology, Centro Hospitalar Universitário do Porto, 4099-001 Porto, Portugal.
| | - Mariana P Monteiro
- Endocrine, Cardiovascular & Metabolic Research, Unit for Multidisciplinary Research in Biomedicine (UMIB), University of Porto, 4050-313 Porto, Portugal.
- Department of Anatomy, Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
38
|
Mei Y, Yang JP, Lang YH, Peng LX, Yang MM, Liu Q, Meng DF, Zheng LS, Qiang YY, Xu L, Li CZ, Wei WW, Niu T, Peng XS, Yang Q, Lin F, Hu H, Xu HF, Huang BJ, Wang LJ, Qian CN. Global expression profiling and pathway analysis of mouse mammary tumor reveals strain and stage specific dysregulated pathways in breast cancer progression. Cell Cycle 2018; 17:963-973. [PMID: 29712537 DOI: 10.1080/15384101.2018.1442629] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
It is believed that the alteration of tissue microenvironment would affect cancer initiation and progression. However, little is known in terms of the underlying molecular mechanisms that would affect the initiation and progression of breast cancer. In the present study, we use two murine mammary tumor models with different speeds of tumor initiation and progression for whole genome expression profiling to reveal the involved genes and signaling pathways. The pathways regulating PI3K-Akt signaling and Ras signaling were activated in Fvb mice and promoted tumor progression. Contrastingly, the pathways regulating apoptosis and cellular senescence were activated in Fvb.B6 mice and suppressed tumor progression. We identified distinct patterns of oncogenic pathways activation at different stages of breast cancer, and uncovered five oncogenic pathways that were activated in both human and mouse breast cancers. The genes and pathways discovered in our study would be useful information for other researchers and drug development.
Collapse
Affiliation(s)
- Yan Mei
- a Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine , Sun Yat-Sen University Cancer Center , Guangzhou 510060 , China
| | - Jun-Ping Yang
- a Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine , Sun Yat-Sen University Cancer Center , Guangzhou 510060 , China
| | - Yan-Hong Lang
- a Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine , Sun Yat-Sen University Cancer Center , Guangzhou 510060 , China
| | - Li-Xia Peng
- a Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine , Sun Yat-Sen University Cancer Center , Guangzhou 510060 , China
| | - Ming-Ming Yang
- b Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University , Guangzhou 510006 , China
| | - Qing Liu
- b Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University , Guangzhou 510006 , China
| | - Dong-Fang Meng
- a Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine , Sun Yat-Sen University Cancer Center , Guangzhou 510060 , China
| | - Li-Sheng Zheng
- a Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine , Sun Yat-Sen University Cancer Center , Guangzhou 510060 , China
| | - Yuan-Yuan Qiang
- a Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine , Sun Yat-Sen University Cancer Center , Guangzhou 510060 , China
| | - Liang Xu
- a Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine , Sun Yat-Sen University Cancer Center , Guangzhou 510060 , China
| | - Chang-Zhi Li
- a Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine , Sun Yat-Sen University Cancer Center , Guangzhou 510060 , China
| | - Wen-Wen Wei
- a Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine , Sun Yat-Sen University Cancer Center , Guangzhou 510060 , China
| | - Ting Niu
- b Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University , Guangzhou 510006 , China
| | - Xing-Si Peng
- a Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine , Sun Yat-Sen University Cancer Center , Guangzhou 510060 , China
| | - Qin Yang
- a Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine , Sun Yat-Sen University Cancer Center , Guangzhou 510060 , China
| | - Fen Lin
- a Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine , Sun Yat-Sen University Cancer Center , Guangzhou 510060 , China
| | - Hao Hu
- d Department of Traditional Chinese Medicine , First Affiliated Hospital, Sun Yat-Sen University , Guangzhou , China
| | - Hong-Fa Xu
- e Department of Hematology , The First Affiliated Hospital of Guangzhou Medical University , Guangzhou 510230 , China
| | - Bi-Jun Huang
- a Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine , Sun Yat-Sen University Cancer Center , Guangzhou 510060 , China
| | - Li-Jing Wang
- b Vascular Biology Research Institute, School of Basic Course, Guangdong Pharmaceutical University , Guangzhou 510006 , China
| | - Chao-Nan Qian
- a Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine , Sun Yat-Sen University Cancer Center , Guangzhou 510060 , China.,c Department of Nasopharyngeal Carcinoma , Sun Yat-Sen University Cancer Center , Guangzhou 510060 , China
| |
Collapse
|
39
|
Akarca-Dizakar SO, Aktuğ H, Oltulu F, Öktem G, Yavaşoğlu A, Açikgöz E, Yiğittürk G, Demir K, Uysal A. Effects of sunitinib on immunoreactivity of vimentin, E-cadherin and S100 in kidneys of streptozotocin induced diabetic mice. Biotech Histochem 2018; 93:328-335. [PMID: 29652183 DOI: 10.1080/10520295.2018.1439532] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Abstract
Diabetes mellitus (DM) affects many organs including kidney. Tyrosine kinase can cause hypoglycemia and sunitinib is an inhibitor of tyrosine kinase. We investigated the possible effects of sunitinib on the kidney of streptozotocin (STZ) induced type 1 diabetic mice. We used 28 CD 1 type male mice divided into four groups of seven. Type 1 diabetes was induced by injection of STZ. Group 1 was the untreated control. Group 2 comprised non-diabetic mice + sunitinib. Both groups 1 and 2 exhibited normal blood glucose levels. Group 3 comprised STZ treated diabetic mice + saline. Group 4 were diabetic mice + sunitinib treatment. Kidneys were removed after 8 weeks. The immunoreactivities of vimentin, E-cadherin and S100 were assessed. Immunostaining of vimentin, E-cadherin and S100 was located in both the glomeruli and tubules of the kidney. We found that the number of vimentin and E-cadherin positive glomeruli and tubules were increased after sunitinib treatment compared to saline treated diabetic mice. The number of vimentin labeled tubules was decreased in the sunitinib treated group compared to diabetic + saline groups. Differences in the number of S100 positive tubules and glomeruli between groups 3 and 4 were not statistically significant. The effect of sunitinib on experimental diabetic mice appears to be related to levels of vimentin, E-cadherin and S100 in the glomeruli and tubules of the kidney, and sunitinib may protect against renal damage from DM.
Collapse
Affiliation(s)
- S O Akarca-Dizakar
- a Department of Histology and Embryology , Gazi University Medical Faculty , Ankara , Turkey
| | - H Aktuğ
- b Department of Histology and Embryology , Ege University Medical Faculty , İzmir , Turkey
| | - F Oltulu
- b Department of Histology and Embryology , Ege University Medical Faculty , İzmir , Turkey
| | - G Öktem
- b Department of Histology and Embryology , Ege University Medical Faculty , İzmir , Turkey
| | - A Yavaşoğlu
- b Department of Histology and Embryology , Ege University Medical Faculty , İzmir , Turkey
| | - E Açikgöz
- b Department of Histology and Embryology , Ege University Medical Faculty , İzmir , Turkey
| | - G Yiğittürk
- b Department of Histology and Embryology , Ege University Medical Faculty , İzmir , Turkey
| | - K Demir
- b Department of Histology and Embryology , Ege University Medical Faculty , İzmir , Turkey
| | - A Uysal
- b Department of Histology and Embryology , Ege University Medical Faculty , İzmir , Turkey
| |
Collapse
|
40
|
Tuo Y, Xiang M. mTOR: A double‐edged sword for diabetes. J Leukoc Biol 2018; 106:385-395. [DOI: 10.1002/jlb.3mr0317-095rr] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 09/05/2017] [Accepted: 09/14/2017] [Indexed: 12/14/2022] Open
Affiliation(s)
- Yali Tuo
- Department of PharmacologySchool of PharmacyTongji Medical CollegeHuazhong University of Science and Technology Wuhan China
| | - Ming Xiang
- Department of PharmacologySchool of PharmacyTongji Medical CollegeHuazhong University of Science and Technology Wuhan China
| |
Collapse
|
41
|
Ng HS, Koczwara B, Roder D, Vitry A. Changes in the prevalence of comorbidity in the Australian population with cancer, 2007-2014. Cancer Epidemiol 2018; 54:56-62. [PMID: 29597133 DOI: 10.1016/j.canep.2018.03.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 03/14/2018] [Accepted: 03/16/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Coexistence of multiple chronic diseases is highly prevalent among the cancer population. This study aims to assess changes in the prevalence of chronic conditions among the population with cancer compared to the Australian general population between 2007 and 2014. METHODS Data from three successive National Health Surveys conducted by the Australian Bureau of Statistics between 2007 and 2014 were utilized. Comparisons were made between the samples of the Australian population aged 25 years and above with a history of cancer and those respondents who did not report having had a cancer using logistics regression models. RESULTS People with a history of cancer had significantly higher odds of reporting non-infectious comorbidity compared to the non-cancer groups across the three surveys. There were no significant changes in the prevalence of diseases affecting circulatory, musculoskeletal, digestive, nervous system, blood and blood forming organs, eye, skin and infectious and parasitic diseases over time among the population with cancer. The prevalence of mental and behavioural problems, endocrine, nutritional and metabolic diseases, and diseases of respiratory and genitourinary system has increased over time among the cancer survivors. CONCLUSION Comorbidity is more prevalent among the cancer population than the general population without cancer. The prevalence of comorbidity was fairly stable for most but not all comorbidities in the population with cancer over the eight-year study period. Further studies on the impacts of coordinated care models for the management of multi-morbidity experienced by cancer survivors that align with the 'National Strategic Framework for Chronic Conditions' are needed.
Collapse
Affiliation(s)
- Huah Shin Ng
- School of Pharmacy and Medical Sciences, University of South Australia, Australia.
| | - Bogda Koczwara
- Flinders Centre for Innovation in Cancer, Flinders University, Australia
| | - David Roder
- Cancer Epidemiology and Population Health, Centre of Population Health Research, School of Health Sciences, University of South Australia, Australia
| | - Agnes Vitry
- School of Pharmacy and Medical Sciences, University of South Australia, Australia
| |
Collapse
|
42
|
Ginsenosides synergize with mitomycin C in combating human non-small cell lung cancer by repressing Rad51-mediated DNA repair. Acta Pharmacol Sin 2018; 39:449-458. [PMID: 28836581 DOI: 10.1038/aps.2017.53] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 04/05/2017] [Indexed: 02/06/2023]
Abstract
The use of ginseng extract as an adjuvant for cancer treatment has been reported in both animal models and clinical applications, but its molecular mechanisms have not been fully elucidated. Mitomycin C (MMC), an anticancer antibiotic used as a first- or second-line regimen in the treatment for non-small cell lung carcinoma (NSCLC), causes serious adverse reactions when used alone. Here, by using both in vitro and in vivo experiments, we provide evidence for an optimal therapy for NSCLC with total ginsenosides extract (TGS), which significantly enhanced the MMC-induced cytotoxicity against NSCLC A549 and PC-9 cells in vitro when used in combination with relatively low concentrations of MMC. A NSCLC xenograft mouse model was used to confirm the in vivo synergistic effects of the combination of TGS with MMC. Further investigation revealed that TGS could significantly reverse MMC-induced S-phase cell cycle arrest and inhibit Rad51-mediated DNA damage repair, which was evidenced by the inhibitory effects of TGS on the levels of phospho-MEK1/2, phospho-ERK1/2 and Rad51 protein and the translocation of Rad51 from the cytoplasm to the nucleus in response to MMC. In summary, our results demonstrate that TGS could effectively enhance the cytotoxicity of MMC against NSCLC cells in vitro and in vivo, thereby revealing a novel adjuvant anticancer mechanism of TGS. Combined treatment with TGS and MMC can significantly lower the required concentration of MMC and can further reduce the risk of side effects, suggesting a better treatment option for NSCLC patients.
Collapse
|
43
|
Worndl E, Fung K, Fischer HD, Austin PC, Krzyzanowska MK, Lipscombe LL. Preventable Diabetic Complications After a Cancer Diagnosis in Patients With Diabetes: A Population-Based Cohort Study. JNCI Cancer Spectr 2018; 2:pky008. [PMID: 29795802 PMCID: PMC5954615 DOI: 10.1093/jncics/pky008] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/12/2018] [Accepted: 03/01/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND A cancer diagnosis may disrupt diabetes management, increasing the risk of preventable complications. The objective was to determine whether a cancer diagnosis in patients with diabetes is associated with an increased risk of diabetic complications. METHODS This retrospective cohort study using health care data from Ontario, Canada, included persons age 50 years or older diagnosed with diabetes from 2007 to 2011 and followed until 2014. We examined the effects of cancer as a time-varying covariate: breast cancer (in women), prostate cancer (in men), colorectal cancer, and other cancers (in men and women). Each cancer exposure was categorized as stage I-III, IV, or unknown, and by time since cancer diagnosis (0-1 year, >1-3 years, and >3 years). The primary outcome was hospital visits for diabetic emergencies. Secondary outcomes were hospital visits for skin and soft tissue infections and cardiovascular events. RESULTS Of 817 060 patients with diabetes (mean age = 64.9 +/- 10.7 years), there were 9759 (1.2%) colorectal and 45 705 (5.6%) other cancers, 6714 (1.7%) breast cancers among 384 257 women and 10 331 (2.4%) prostate cancers among 432 803 men. For all cancers except stage I-III prostate cancer, rates of diabetic complications were significantly higher zero years to one year after diagnosis compared with no cancer (adjusted relative rates ranging from 1.26, 95% confidence interval [CI] = 1.08 to 1.49, to 4.07, 95% CI = 3.80 to 4.36); these differences were attenuated in the subsequent periods after cancer diagnosis. CONCLUSIONS Patients with diabetes are at increased risk for preventable complications after a cancer diagnosis. Better diabetes care is needed during this vulnerable period.
Collapse
Affiliation(s)
- Erin Worndl
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Kinwah Fung
- Institute for Clinical Evaluative Sciences, University of Toronto, Toronto, ON, Canada
| | - Hadas D Fischer
- Institute for Clinical Evaluative Sciences, University of Toronto, Toronto, ON, Canada
| | - Peter C Austin
- Institute for Clinical Evaluative Sciences, University of Toronto, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
| | - Monika K Krzyzanowska
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Institute for Clinical Evaluative Sciences, University of Toronto, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
- Department of Medical Oncology, Princess Margaret Hospital, Toronto, ON, Canada
| | - Lorraine L Lipscombe
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Institute for Clinical Evaluative Sciences, University of Toronto, Toronto, ON, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, ON, Canada
- Department of Medicine, Women’s College Research Institute, Women’s College Hospital, Toronto, ON, Canada
| |
Collapse
|
44
|
Wang J, Zhao Y, Wang Q, Zhang L, Shi J, Wang Z, Cheng Y, He J, Shi Y, Yu H, Zhao Y, Chen W, Luo Y, Wang X, Nan K, Jin F, Dong J, Li B, Liu Z, Han B, Li K. Prognostic factors of refractory NSCLC patients receiving anlotinib hydrochloride as the third- or further-line treatment. Cancer Biol Med 2018; 15:443-451. [PMID: 30766754 PMCID: PMC6372914 DOI: 10.20892/j.issn.2095-3941.2018.0158] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Objective: Anlotinib hydrochloride is a multitarget tyrosine kinase inhibitor that targets vascular endothelial growth factor receptor, fibroblast growth factor receptor, platelet-derived growth factor receptor, c-Kit, and c-MET; therefore, it exhibits both antitumor and anti-angiogenetic activities. A phase III trial has shown that anlotinib improved progression-free survival (PFS) and overall survival (OS) in patients with advanced non-small cell lung cancer (NSCLC), who presented with progressive disease or intolerance after standard chemotherapy. This study aimed to analyze the characteristics of patients receiving anlotinib treatment to determine the dominant populations who are fit for the treatment. Methods: Data were collected from March 2015 to January 2017 from a randomized, double-blind, placebo-controlled, multicenter, phase III trial of anlotinib (ALTER0303). A total of 437 patients were enrolled and randomly allocated (2:1) to the anlotinib and placebo groups. Kaplan–Meier analysis and log-rank test were performed to compare PFS and OS. Cox proportional hazards model was adopted for multivariate prognostic analysis. Results: Multivariate analysis indicated that high post-therapeutic peripheral blood granulocyte/lymphocyte ratio and elevated alkaline phosphatase levels were independent risk factors for PFS. Meanwhile, elevated thyroid-stimulating hormone, blood glucose, and triglyceride levels; hypertension; and hand–foot syndrome were independent protective factors of PFS. High post-therapeutic peripheral blood granulocyte/lymphocyte ratio, an Eastern Cooperative Oncology Group (ECOG) score ≥ 2, and the sum of the maximal target lesion length at baseline were independent risk factors of OS, and hypertriglyceridemia was an independent protective factor of OS. Conclusions: This study preliminarily explored the possible factors that affected PFS and OS after anlotinib treatment in patients with advanced refractory NSCLC, and the baseline characteristics of the therapeutically dominant populations were then identified.
Collapse
Affiliation(s)
- Jing Wang
- Department of Pulmonary Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Yizhuo Zhao
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai 230030, China
| | - Qiming Wang
- Department of Medical Oncology, Henan Province Tumor Hospital, Zhengzhou 450008, China
| | - Li Zhang
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Beijing 100730 China
| | - Jianhua Shi
- Department of Medical Oncology, Linyi Cancer Hospital, Linyi 276001, China
| | - Zhehai Wang
- Department of Internal Medicine, Shandong Cancer Hospital, Jinan 250117, China
| | - Ying Cheng
- Department of Thoracic Oncology, Jilin Cancer Hospital, Changchun 130012, China
| | - Jianxing He
- Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Yuankai Shi
- Department of Medical Oncology, Cancer Hospital Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Hao Yu
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Yang Zhao
- Department of Biostatistics, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Weiqiang Chen
- Department of Respiratory Medicine, Lanzhou Military General Hospital, Lanzhou 730050, China
| | - Yi Luo
- Department of Head and Neck Oncology, Hunan Cancer Hospital, Changsha 220633, China
| | - Xiuwen Wang
- Department of Chemotherapy, Qilu Hospital of Shandong University, Jinan 250000, China
| | - Kejun Nan
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China
| | - Faguang Jin
- Department of Respiratory and Critical Diseases, Tang Du Hospital, Xi'an 710038, China
| | - Jian Dong
- Department of Oncology, Yunnan Cancer Hospital, Kunming 650032, China
| | - Baolan Li
- General Department, Capital Medical University Beijing Chest Hospital, Beijing 101149, China
| | - Zhujun Liu
- Department of Pulmonary Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Baohui Han
- Department of Respiratory Medicine, Shanghai Chest Hospital, Shanghai 230030, China
| | - Kai Li
- Department of Pulmonary Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| |
Collapse
|
45
|
Zanders MMJ, Haak HR, van Herk-Sukel MPP, Herings RMC, van de Poll-Franse LV, Johnson JA. Changes in glucose-lowering drug use before and after cancer diagnosis in patients with diabetes. DIABETES & METABOLISM 2017; 44:22-29. [PMID: 29066209 DOI: 10.1016/j.diabet.2017.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 08/11/2017] [Accepted: 08/28/2017] [Indexed: 12/18/2022]
Abstract
AIM This study explores the changes in glucose-lowering drug (GLD) use before and after cancer diagnosis among patients with diabetes. METHODS New GLD users (1998-2011) living in the Dutch ECR-PHARMO catchment area were selected from the PHARMO Database Network (n=52,228). Those with a primary cancer diagnosis were considered cases (n=3281) and matched with eligible controls (n=12,891) without cancer during follow-up. Conditional logistic regression analysis was used to assess changes in GLD use, such as treatment add-ons, treatments drops and initiation of insulin, for cases compared with controls associated with specific cancer types in four time windows (6-3 and 0-3months before cancer diagnosis; 0-3 and 3-6months after cancer diagnosis). RESULTS In the 3months before cancer diagnosis, patients with upper gastrointestinal (GI) cancers (oesophageal, stomach, pancreatic, liver cancers) had higher odds of initiating insulin (OR: 9.3; 95% CI: 3.6-24.1); to a lesser extent, this was also observed in the 3months prior to that (at 6months, OR: 3.9; 95% CI: 1.3-12.1). Diagnosis of colorectal (OR: 3.4; 95% CI: 1.4-8.4), pulmonary (OR: 2.5; 95% CI: 1.1-5.4) and upper GI (OR: 13.6; 95% CI: 5.0-36.9) cancers was associated with increased odds of initiating insulin in the 3months after cancer diagnosis. During all study time windows, the odds of treatment drops were higher for patients with upper GI cancers whereas, for most other cancers, these odds were higher only after a diagnosis of cancer. CONCLUSION The greater odds of initiating insulin during the 6months prior to diagnosis of upper GI cancers suggest reverse causation. After cancer diagnosis, drops in use of GLDs was commonly seen.
Collapse
Affiliation(s)
- M M J Zanders
- Netherlands Comprehensive Cancer Organisation,, P.O. Box 231, 5600 AE Eindhoven, The Netherlands; Department of Internal Medicine, Máxima Medical Centre, Eindhoven/Veldhoven, P.O. Box 7777. 5500 MB Veldhoven, The Netherlands.
| | - H R Haak
- Department of Internal Medicine, Máxima Medical Centre, Eindhoven/Veldhoven, P.O. Box 7777. 5500 MB Veldhoven, The Netherlands; Department of Internal Medicine, Division of General Internal Medicine, Maastricht University Medical Centre+, P.O. Box 5800, 6229 HX, Maastricht, The Netherlands; Maastricht University, Department of Health Services Research, and CAPHRI School for Public Health and Primary Care, P.O. Box 616, Maastricht, 6200 MD, The Netherlands
| | - M P P van Herk-Sukel
- PHARMO Institute for Drug Outcomes Research, Van Deventerlaan 30-40, 3528 AE, Utrecht, The Netherlands
| | - R M C Herings
- PHARMO Institute for Drug Outcomes Research, Van Deventerlaan 30-40, 3528 AE, Utrecht, The Netherlands
| | - L V van de Poll-Franse
- Netherlands Comprehensive Cancer Organisation,, P.O. Box 231, 5600 AE Eindhoven, The Netherlands; Center of Research on Psychology in Somatic Diseases (CoRPS), Department of Medical and Clinical Psychology, Tilburg University, P.O. Box 90153, 5000 LE, Tilburg, The Netherlands; Division of Psychosocial Oncology and Epidemiology, Netherlands Cancer Institute, Amsterdam, P.O. Box 90203, 1006 BE, Amsterdam, The Netherlands
| | - J A Johnson
- School of Public Health, University of Alberta, 87 Ave, 11405 Edmonton, AB T6G 1C9, Canada
| |
Collapse
|
46
|
Martins D, Lambrescu I, Barucca V, Fazio N. Everolimus-related adverse events in neuroendocrine tumors and comparative considerations with breast and renal cancer: a critical overview. Expert Opin Orphan Drugs 2017. [DOI: 10.1080/21678707.2017.1334550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Diana Martins
- Endocrinology, Diabetes and Metabolism Department of Coimbra Hospital and University Centre, Coimbra, Portugal
| | - Ioana Lambrescu
- Endocrinology Department, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - Viola Barucca
- Division of Medical Oncology, Department of Oncology, Misericordia General Hospital, Grosseto, Italy
| | - Nicola Fazio
- Unit of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of oncology, IEO, Milan, Italy
| |
Collapse
|
47
|
Prediction of Everolimus Toxicity and Prognostic Value of Skeletal Muscle Index in Patients With Metastatic Renal Cell Carcinoma. Clin Genitourin Cancer 2017; 15:350-355. [DOI: 10.1016/j.clgc.2017.01.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 01/16/2017] [Accepted: 01/23/2017] [Indexed: 01/15/2023]
|
48
|
Lin M, Jin J. Cancer, obesity, and diabetes: TKIs exert multiple effects on glucose homeostasis. Nat Rev Clin Oncol 2017; 14:268. [PMID: 28422115 DOI: 10.1038/nrclinonc.2017.56] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Minglin Lin
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, Guangxi, China
| | - Junfei Jin
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, 15 Lequn Road, Guilin, 541001, Guangxi, China
| |
Collapse
|
49
|
Gutiérrez-Salmerón M, Chocarro-Calvo A, García-Martínez JM, de la Vieja A, García-Jiménez C. Epidemiological bases and molecular mechanisms linking obesity, diabetes, and cancer. ACTA ACUST UNITED AC 2017; 64:109-117. [PMID: 28440775 DOI: 10.1016/j.endinu.2016.10.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 09/29/2016] [Accepted: 10/10/2016] [Indexed: 02/07/2023]
Abstract
The association between diabetes and cancer was hypothesized almost one century ago. Today, a vast number of epidemiological studies support that obese and diabetic populations are more likely to experience tissue-specific cancers, but the underlying molecular mechanisms remain unknown. Obesity, diabetes, and cancer share many hormonal, immune, and metabolic changes that may account for the relationship between diabetes and cancer. In addition, antidiabetic treatments may have an impact on the occurrence and course of some cancers. Moreover, some anticancer treatments may induce diabetes. These observations aroused a great controversy because of the ethical implications and the associated commercial interests. We report an epidemiological update from a mechanistic perspective that suggests the existence of many common and differential individual mechanisms linking obesity and type 1 and 2 diabetes mellitus to certain cancers. The challenge today is to identify the molecular links responsible for this association. Classification of cancers by their molecular signatures may facilitate future mechanistic and epidemiological studies.
Collapse
Affiliation(s)
- María Gutiérrez-Salmerón
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Madrid, España
| | - Ana Chocarro-Calvo
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Madrid, España; Nuffield Department of Clinical Medicine, Ludwig Institute for Cancer Research, Universidad de Oxford, Headington, Oxford, Reino Unido
| | - José Manuel García-Martínez
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Madrid, España
| | - Antonio de la Vieja
- Unidad de Tumores Endocrinos (UFIEC), Instituto de Salud Carlos III, Majadahonda, Madrid, España.
| | - Custodia García-Jiménez
- Departamento de Ciencias Básicas de la Salud, Facultad de Ciencias de la Salud, Universidad Rey Juan Carlos, Alcorcón, Madrid, España.
| |
Collapse
|
50
|
Romero-Ventosa EY, Otero-Millán L, González-Costas S, Vilasoa-Boo P, Silva-López A, Arroyo-Conde C, Piñeiro-Corrales G. Effect of tyrosine kinase inhibitors on the glucose levels in diabetic and nondiabetic patients. Indian J Cancer 2017; 54:136-143. [DOI: 10.4103/ijc.ijc_190_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|