1
|
Chen J, Xia D, Ke L. Anlotinib induced type 1 diabetes: a case report. Front Oncol 2025; 15:1508645. [PMID: 40242243 PMCID: PMC11999947 DOI: 10.3389/fonc.2025.1508645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 03/19/2025] [Indexed: 04/18/2025] Open
Abstract
Studies have shown some tyrosine kinase inhibitors (TKIs) can influence glucose metabolism leading to either hypoglycemia or hyperglycemia which is reversable in most patients after treatment cessation. Anlotinib is a novel oral multi-target tyrosine kinase inhibitor (TKI) which has been approved for non-small cell lung cancer in China. Previous studies of anlotinib did not report it has any side effect on blood glucose, and there has been no case reporting type 1 diabetes associated with any TKI. The present case study, to our knowledge, was the first to report on an 81-year-old man with lung cancer who developed type 1 diabetes following 14 cycles treatment with TKI. The fasting plasma blood glucose and hemoglobinA1c (HbA1c) was 24.3mmol/L and 9.0%, respectively, and GADA (glutamic acid decarboxylase antibody) was more than 2000IU/ml (normal range is less than 10IU/ml) when he was diagnosed. We also conducted a literature review to explore the potential mechanism of anlotinib in inducing type 1 diabetes and recommend that self-monitoring blood glucose (SMBG) for fasting and random postprandial blood glucose at least once a week is needed for early identification of glucose dysregulation when using TKI drugs, and monthly fasting and random postprandial plasma glucose monitoring and HbA1c test every 3 months is also recommended if the SMBG protocol cannot be completed.
Collapse
Affiliation(s)
| | | | - Linqiu Ke
- General Medicine Department, Chongqing General Hospital, Chongqing, China
| |
Collapse
|
2
|
Indarwulan N, Savitri M, Ashariati A, Bintoro SUY, Diansyah MN, Amrita PNA, Romadhon PZ. Bone Mineral Density, C-Terminal Telopeptide of Type I Collagen, and Osteocalcin as Monitoring Parameters of Bone Remodeling in CML Patients Undergoing Imatinib Therapy: A Basic Science and Clinical Review. Diseases 2024; 12:275. [PMID: 39589949 PMCID: PMC11592756 DOI: 10.3390/diseases12110275] [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: 09/17/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Chronic myeloid leukemia (CML) is one of the most commonly found types of myeloproliferative neoplasms, characterized by increased proliferation of granulocytic cells without losing their differentiation ability. Imatinib, a tyrosine kinase inhibitor (TKI), can be effectively used as therapy for CML. However, Imatinib can affect bone turnover thus having clinical implications on the bones of CML patients undergoing long-term Imatinib therapy. However, parameters that can accurately describe the bone condition in CML patients receiving Imatinib still need further study. A combination of imaging techniques such as bone mineral density (BMD) and bone turnover activity markers such as C-terminal telopeptide of type I collagen (CTX-1) and osteocalcin has the potential to be used as monitoring parameters for bone density abnormalities in CML patients receiving Imatinib. OBJECTIVES This article explains the rationale for using BMD, CTX-1, and osteocalcin as monitoring parameters of bone remodeling in CML patients receiving Imatinib. RESULTS First, the physiological process of bone turnover will be explained. Then, we describe the role of tyrosine kinase in bone metabolism. Next, the impact of Imatinib on BMD, CTX-1, and osteocalcin will be explained. CONCLUSION The assessment of bone health of CML patients on Imatinib should include both BMD tests and bone turnover marker assays such as CTX-1 and osteocalcin.
Collapse
Affiliation(s)
- Nurita Indarwulan
- Subspeciality Program in Hematology and Medical Oncology Division, Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia;
- Subspeciality Program in Hematology and Medical Oncology Division, Department of Internal Medicine, Faculty of Medicine, Airlangga University, Surabaya 60132, Indonesia
| | - Merlyna Savitri
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia; (A.A.); (S.U.Y.B.); (M.N.D.); (P.N.A.A.); (P.Z.R.)
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Airlangga University, Surabaya 60132, Indonesia
| | - Ami Ashariati
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia; (A.A.); (S.U.Y.B.); (M.N.D.); (P.N.A.A.); (P.Z.R.)
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Airlangga University, Surabaya 60132, Indonesia
| | - Siprianus Ugroseno Yudho Bintoro
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia; (A.A.); (S.U.Y.B.); (M.N.D.); (P.N.A.A.); (P.Z.R.)
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Airlangga University, Surabaya 60132, Indonesia
| | - Muhammad Noor Diansyah
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia; (A.A.); (S.U.Y.B.); (M.N.D.); (P.N.A.A.); (P.Z.R.)
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Airlangga University, Surabaya 60132, Indonesia
| | - Putu Niken Ayu Amrita
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia; (A.A.); (S.U.Y.B.); (M.N.D.); (P.N.A.A.); (P.Z.R.)
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Airlangga University, Surabaya 60132, Indonesia
| | - Pradana Zaky Romadhon
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Dr. Soetomo General Academic Hospital, Surabaya 60286, Indonesia; (A.A.); (S.U.Y.B.); (M.N.D.); (P.N.A.A.); (P.Z.R.)
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Faculty of Medicine, Airlangga University, Surabaya 60132, Indonesia
| |
Collapse
|
3
|
Tomuleasa C, Tigu AB, Munteanu R, Moldovan CS, Kegyes D, Onaciu A, Gulei D, Ghiaur G, Einsele H, Croce CM. Therapeutic advances of targeting receptor tyrosine kinases in cancer. Signal Transduct Target Ther 2024; 9:201. [PMID: 39138146 PMCID: PMC11323831 DOI: 10.1038/s41392-024-01899-w] [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: 01/19/2024] [Revised: 05/29/2024] [Accepted: 06/14/2024] [Indexed: 08/15/2024] Open
Abstract
Receptor tyrosine kinases (RTKs), a category of transmembrane receptors, have gained significant clinical attention in oncology due to their central role in cancer pathogenesis. Genetic alterations, including mutations, amplifications, and overexpression of certain RTKs, are critical in creating environments conducive to tumor development. Following their discovery, extensive research has revealed how RTK dysregulation contributes to oncogenesis, with many cancer subtypes showing dependency on aberrant RTK signaling for their proliferation, survival and progression. These findings paved the way for targeted therapies that aim to inhibit crucial biological pathways in cancer. As a result, RTKs have emerged as primary targets in anticancer therapeutic development. Over the past two decades, this has led to the synthesis and clinical validation of numerous small molecule tyrosine kinase inhibitors (TKIs), now effectively utilized in treating various cancer types. In this manuscript we aim to provide a comprehensive understanding of the RTKs in the context of cancer. We explored the various alterations and overexpression of specific receptors across different malignancies, with special attention dedicated to the examination of current RTK inhibitors, highlighting their role as potential targeted therapies. By integrating the latest research findings and clinical evidence, we seek to elucidate the pivotal role of RTKs in cancer biology and the therapeutic efficacy of RTK inhibition with promising treatment outcomes.
Collapse
Affiliation(s)
- Ciprian Tomuleasa
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania.
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania.
| | - Adrian-Bogdan Tigu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Raluca Munteanu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Cristian-Silviu Moldovan
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - David Kegyes
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Academy of Romanian Scientists, Ilfov 3, 050044, Bucharest, Romania
| | - Anca Onaciu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Diana Gulei
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Gabriel Ghiaur
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Department of Leukemia, Sidney Kimmel Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hermann Einsele
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
- Universitätsklinikum Würzburg, Medizinische Klinik II, Würzburg, Germany
| | - Carlo M Croce
- Department of Cancer Biology and Genetics and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
4
|
Gendron LN, Sheveland CG, Gunn JR, Pogue BW, Shell TA, Shell JR. Radiation-Activated Cobalamin-Kinase Inhibitors for Treatment of Pancreatic Ductal Adenocarcinoma. Mol Pharm 2024; 21:137-142. [PMID: 37989273 PMCID: PMC11228961 DOI: 10.1021/acs.molpharmaceut.3c00667] [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] [Indexed: 11/23/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains one of the most dismal diagnoses that a patient can receive. PDAC is extremely difficult to treat, as drug delivery is challenging in part due to the lack of vascularization, high stromal content, and high collagen content of these tumors. We have previously demonstrated that attaching drugs to the cobalamin scaffold provides selectivity for tumors over benign cells due to a high vitamin demand in these rapidly growing cells and an overexpression of transcobalamin receptors in a variety of cancer types. Importantly, we have shown the ability to deliver cobalamin derivatives to orthotopic pancreas tumors. Tyrosine kinase inhibitors have shown promise in treating PDAC as well as other cancer types. However, some of these inhibitors suffer from drug resistance, and as such, their success has been diminished. With this in mind, we synthesized the tyrosine kinase inhibitors erlotinib (EGFR) and dasatinib (Src) that are attached to this cobalamin platform. Both of these cobalamin-drug conjugates cause visible light-induced apoptosis, and the cobalamin-erlotinib conjugate (2) causes X-ray-induced apoptosis in MIA PaCa-2 cells. Both visible light and X-rays provide spatial control of drug release; however, utilizing X-ray irradiation offers the advantage of deeper tissue penetration. Therefore, we explored the utilization of 2 as a synergistic therapy with radiation in athymic nude mice implanted with MIA PaCa-2 tumors. We discovered that the addition of 2 caused an enhanced reduction in tumor margins in comparison with radiation therapy alone. In addition, treatment with 2 in the absence of radiation caused no significant reduction in tumor size in comparison with the controls. The cobalamin technology presented here allows for the spatial release of drugs in conjunction with external beam radiation therapy, potentially allowing for more effective treatment of deep-seated tumors with less systemic side effects.
Collapse
Affiliation(s)
- Liberty N Gendron
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
| | - Colter G Sheveland
- Department of Chemistry and Biochemistry, Norwich University, Northfield, Vermont 05663, United States
| | - Jason R Gunn
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
| | - Brian W Pogue
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
- Department of Medical Physics, University of Wisconsin, Madison, Wisconsin 53705, United States
| | - Thomas A Shell
- Department of Chemistry and Physics, Lincoln Memorial University, Harrogate, Tennessee 37752, United States
| | - Jennifer R Shell
- Thayer School of Engineering, Dartmouth College, Hanover, New Hampshire 03755, United States
- Eos Pharmaceuticals LLC, Tazewell, Tennessee 37879, United States
| |
Collapse
|
5
|
Sontag I, Bergmann L, Adamek HE. Severe Hyperglycemia Due to Protein Kinase Inhibitor Therapy in a Patient With Poorly Controlled Diabetes Mellitus. JCEM CASE REPORTS 2024; 2:luad172. [PMID: 38188906 PMCID: PMC10768878 DOI: 10.1210/jcemcr/luad172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Indexed: 01/09/2024]
Abstract
The efficacy and safety of zanubrutinib, a highly selective next-generation Bruton's tyrosine kinase (BTK) inhibitor, in chronic lymphocytic leukemia and lymphoplasmocytoides immunocytoma seems favorable. Adverse events comprise neutropenia, thrombocytopenia, infection, anemia, and atrial fibrillation. This report describes a 75-year-old man suffering from polydipsia, polyuria, and blurred vision for 10 days. He was diagnosed with lymphoplasmocytoides immunocytoma in 2003. After various therapies, he was started on zanubrutinib in October 2022. A diagnosis of diabetes mellitus had never been established before. On arrival in the emergency department, his plasma glucose was 37.2 mmol/L (671 mg/dL) and glycated hemoglobin (HbA1c) was 14.2%. Circulating antibodies showed positivity for glutamic acid decarboxylase (GAD-65), and his C-peptide level was 1.3 nmol/L (normal range, 0.37-1.47 nmol/L), equivalent to 3.9 ng/mL (normal range 1.1-5.0 ng/mL). From the patient's medical history, it became obvious that the metabolic situation had been problematic for many years, and that diabetes could have been taken into account at least in the summer of 2020 when HbA1c was 6.7%. In patients on tyrosine kinase inhibitors, careful assessment of glycemic control (monitoring HbA1c and blood glucose levels periodically even for nondiabetic patients) is recommended to prevent a major diabetic emergency.
Collapse
Affiliation(s)
- Isabel Sontag
- Zentrale Notfallambulanz, Klinikum Leverkusen, Academic Hospital of the University of Cologne, Am Gesundheitspark 11, D—51375 Leverkusen, Germany
| | - Laura Bergmann
- Med. Klinik 2, Klinikum Leverkusen, Academic Hospital of the University of Cologne, Am Gesundheitspark 11, D—51375 Leverkusen, Germany
| | - Henning Ernst Adamek
- Med. Klinik 2, Klinikum Leverkusen, Academic Hospital of the University of Cologne, Am Gesundheitspark 11, D—51375 Leverkusen, Germany
| |
Collapse
|
6
|
Shyam Sunder S, Sharma UC, Pokharel S. Adverse effects of tyrosine kinase inhibitors in cancer therapy: pathophysiology, mechanisms and clinical management. Signal Transduct Target Ther 2023; 8:262. [PMID: 37414756 PMCID: PMC10326056 DOI: 10.1038/s41392-023-01469-6] [Citation(s) in RCA: 151] [Impact Index Per Article: 75.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/06/2023] [Accepted: 04/23/2023] [Indexed: 07/08/2023] Open
Abstract
Since their invention in the early 2000s, tyrosine kinase inhibitors (TKIs) have gained prominence as the most effective pathway-directed anti-cancer agents. TKIs have shown significant utility in the treatment of multiple hematological malignancies and solid tumors, including chronic myelogenous leukemia, non-small cell lung cancers, gastrointestinal stromal tumors, and HER2-positive breast cancers. Given their widespread applications, an increasing frequency of TKI-induced adverse effects has been reported. Although TKIs are known to affect multiple organs in the body including the lungs, liver, gastrointestinal tract, kidneys, thyroid, blood, and skin, cardiac involvement accounts for some of the most serious complications. The most frequently reported cardiovascular side effects range from hypertension, atrial fibrillation, reduced cardiac function, and heart failure to sudden death. The potential mechanisms of these side effects are unclear, leading to critical knowledge gaps in the development of effective therapy and treatment guidelines. There are limited data to infer the best clinical approaches for the early detection and therapeutic modulation of TKI-induced side effects, and universal consensus regarding various management guidelines is yet to be reached. In this state-of-the-art review, we examine multiple pre-clinical and clinical studies and curate evidence on the pathophysiology, mechanisms, and clinical management of these adverse reactions. We expect that this review will provide researchers and allied healthcare providers with the most up-to-date information on the pathophysiology, natural history, risk stratification, and management of emerging TKI-induced side effects in cancer patients.
Collapse
Affiliation(s)
- Sunitha Shyam Sunder
- Cardio-Oncology Research Group, Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Umesh C Sharma
- Division of Cardiovascular Medicine, Jacob's School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Saraswati Pokharel
- Cardio-Oncology Research Group, Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| |
Collapse
|
7
|
Valerio L, Giani C, Matrone A, Pontillo-Contillo B, Minaldi E, Agate L, Molinaro E, Elisei R. Adrenal insufficiency in thyroid cancer patients treated with tyrosine kinase inhibitors and detected by ACTH stimulation test. J Endocrinol Invest 2023:10.1007/s40618-023-02025-3. [PMID: 36809657 DOI: 10.1007/s40618-023-02025-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 01/25/2023] [Indexed: 02/23/2023]
Abstract
PURPOSE Advanced thyroid cancer patients treated with tyrosine kinase inhibitors (TKI) can develop several adverse events (AEs), including adrenal insufficiency (AI). METHODS We studied 55 patients treated with TKI for radioiodine-refractory or medullary thyroid cancer. The adrenal function was evaluated during follow-up by performing serum basal ACTH, and basal and ACTH-stimulated cortisol. RESULTS Twenty-nine/55 (52.7%) patients developed subclinical AI during TKI treatment as demonstrated by a blunted cortisol response to ACTH stimulation. All cases showed normal values of serum sodium, potassium and blood pressure. All patients were immediately treated, and none showed an overt AI. Cases with AI were all negative for adrenal antibodies and did not show any adrenal gland alteration. Other causes of AI were excluded. The onset time of the AI, as measured in the subgroup with a first negative ACTH test, was < 12 months in 5/9 (55.6%), between 12 and 36 months in 2/9 (22.2%) and > 36 months in 2/9 (22.2%) cases. In our series, the only prognostic factor of AI was the elevated, although moderate, basal level of ACTH when the basal and stimulated cortisol were still normal. The glucocorticoid therapy improved fatigue in most patients. CONCLUSIONS Subclinical AI can be developed in > 50% of advanced thyroid cancer patients treated with TKI. This AE can develop in a wide period ranging from < 12 to > 36 months. For this reason, AI must be looked for throughout the follow-up to be early recognized and treated. A periodic ACTH stimulation test, every 6-8 months, can be helpful.
Collapse
Affiliation(s)
- L Valerio
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University of Pisa, Via Paradisa 2, 56124, Pisa, Italy
| | - C Giani
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University of Pisa, Via Paradisa 2, 56124, Pisa, Italy
| | - A Matrone
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University of Pisa, Via Paradisa 2, 56124, Pisa, Italy
| | - B Pontillo-Contillo
- Diagnostic and Interventional Radiology Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Paradisa 2, 56124, Pisa, Italy
| | - E Minaldi
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University of Pisa, Via Paradisa 2, 56124, Pisa, Italy
| | - L Agate
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University of Pisa, Via Paradisa 2, 56124, Pisa, Italy
| | - E Molinaro
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University of Pisa, Via Paradisa 2, 56124, Pisa, Italy
| | - R Elisei
- Department of Clinical and Experimental Medicine, Unit of Endocrinology, University of Pisa, Via Paradisa 2, 56124, Pisa, Italy.
| |
Collapse
|
8
|
VOLTA ALBERTODALLA, DELBARBA ANDREA, VALCAMONICO FRANCESCA, CAPPELLI CARLO, CARAMELLA IRENE, BERGAMINI MARCO, FERLIN ALBERTO, BERRUTI ALFREDO. Gonadal Function in Male Patients With Metastatic Renal Cell Cancer Treated With Sunitinib. In Vivo 2023; 37:410-416. [PMID: 36593059 PMCID: PMC9843777 DOI: 10.21873/invivo.13093] [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: 11/02/2022] [Revised: 11/23/2022] [Accepted: 11/25/2022] [Indexed: 01/04/2023]
Abstract
BACKGROUND/AIM Single-agent tyrosine kinase inhibitors are still prescribed as first-line treatment to a relevant subgroup of patients with metastatic renal cell carcinoma (mRCC). These agents are known to cause disfunction of many endocrine glands (e.g., thyroid). In this two-step trial, we aimed to assess gonadal function among male patients with mRCC treated with sunitinib. PATIENTS AND METHODS We enrolled a first cross-sectional cohort of pre-treated (>6 months) patients and a subsequent cohort of treatment-naïve patients who were prospectively followed-up. All patients were screened for hypogonadism and received a Functional Assessment of Cancer Therapy - General (FACT-G) questionnaire at study entry and after 6 months of therapy. Patients who were candidates for testosterone replacement therapy (TRT) also received a FACT-G questionnaire at baseline and 3 months after supplementation. RESULTS Among the 30 enrolled patients, the prevalence of hypogonadism was found to be higher in those receiving sunitinib for a longer period (27.3% at baseline, 41.7% in the first 6 months, and 68.4% after 9 months of therapy). The testosterone level of patients correlated with quality of life (R=0.32). A total of six patients received TRT, with a significant improvement in their global quality of life after the first 3 months of treatment. CONCLUSION An increasing prevalence of hypogonadism was seen among male patients who received long-term treatment with sunitinib. TRT was associated with relevant improvements in quality of life. These findings corroborate similar published observations and encourage the assessment of gonadal function in male patients with mRCC under treatment with sunitinib.
Collapse
Affiliation(s)
- ALBERTO DALLA VOLTA
- Medical Oncology Unit, ASST Spedali Civili, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - ANDREA DELBARBA
- Unit of Endocrinology and Metabolism, ASST Spedali Civili, Department of Medicine, University of Brescia, Brescia, Italy
| | - FRANCESCA VALCAMONICO
- Medical Oncology Unit, ASST Spedali Civili, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - CARLO CAPPELLI
- Unit of Endocrinology and Metabolism, ASST Spedali Civili, Department of Medicine, University of Brescia, Brescia, Italy
| | - IRENE CARAMELLA
- Medical Oncology Unit, ASST Spedali Civili, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - MARCO BERGAMINI
- Medical Oncology Unit, ASST Spedali Civili, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - ALBERTO FERLIN
- Unit of Endocrinology and Metabolism, ASST Spedali Civili, Department of Medicine, University of Brescia, Brescia, Italy,Unit of Andrology and Reproductive Medicine, Department of Medicine, University of Padua, Padua, Italy
| | - ALFREDO BERRUTI
- Medical Oncology Unit, ASST Spedali Civili, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| |
Collapse
|
9
|
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
|
10
|
Yamauchi I, Sakane Y, Okuno Y, Sugawa T, Hakata T, Fujita H, Okamoto K, Taura D, Yamashita T, Hirota K, Ueda Y, Fujii T, Yasoda A, Inagaki N. High-throughput Screening in Combination With a Cohort Study for Iodothyronine Deiodinases. Endocrinology 2022; 163:6607576. [PMID: 35695766 DOI: 10.1210/endocr/bqac090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Indexed: 11/19/2022]
Abstract
Regulatory mechanisms of iodothyronine deiodinases (DIOs) require further elucidation, and conventional methods for evaluating DIOs are unsuitable for high-throughput screening (HTS). Here we explored factors of transcriptional regulation of 3 types of DIOs (DIO1, DIO2, and DIO3) from a chemical library using our designed HTS. We constructed HTS based on a promoter assay and performed a screen of 2480 bioactive compounds. For compounds that were clinically approved, we validated hit compounds through a retrospective cohort study in our department that evaluated changes in thyroid function in patients using the compounds as drug therapy. Furthermore, we verified the involvement of DIOs using mice treated with the compounds. Of the hit compounds, 6 and 7 compounds transcriptionally up- and downregulated DIO1, respectively; 34 transcriptionally upregulated DIO2; and 5 and 2 compounds transcriptionally up- and downregulated DIO3, respectively. The cohort study clarified the clinical effects of some hit compounds: ritodrine increased free triiodothyronine (fT3)/free thyroxine (fT4) ratio and decreased serum thyroid-stimulating hormone (TSH) levels, tadalafil increased serum fT3 levels, and tyrosine kinase inhibitors (TKIs) decreased serum fT3 and fT4 levels and increased serum TSH levels. Following in vivo experiments using treated mice, consistent results were observed in ritodrine, which upregulated DIO2 in the thyroid gland. In conclusion, we completed HTS for DIOs and obtained attractive hit compounds. Our cohort study revealed the clinical significance of ritodrine, sildenafil, and TKIs. We hope our unique method will contribute to analyzing various targets and lists of hit compounds will promote understanding of DIOs.
Collapse
Affiliation(s)
- Ichiro Yamauchi
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoriko Sakane
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Sugawa Clinic, Kyoto, Japan
| | - Yukiko Okuno
- Medical Research Support Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Taku Sugawa
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takuro Hakata
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Haruka Fujita
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kentaro Okamoto
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Daisuke Taura
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takafumi Yamashita
- Metabolism and Endocrinology Division of Internal Medicine, Kishiwada City Hospital, Osaka, Japan
| | - Keisho Hirota
- Department of Pathology and Biology of Diseases, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yohei Ueda
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Toshihito Fujii
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Akihiro Yasoda
- Clinical Research Center, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Nobuya Inagaki
- Department of Diabetes, Endocrinology and Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
11
|
Endocrine Late Effects in Childhood Cancer Survivors. Cancers (Basel) 2022; 14:cancers14112630. [PMID: 35681610 PMCID: PMC9179858 DOI: 10.3390/cancers14112630] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Recent advances in cancer treatment have led to improved survival, with an exponential increase in sequelae among survivors. Around 50% of survivors will experience at least one hormonal disorder, with radiotherapy, hematopoietic stem cell transplantation, and alkylating chemotherapy being the most frequently related. Therefore, lifelong monitoring of childhood cancer survivors at risk is paramount. With this review, we describe in detail the most prevalent endocrine sequelae, considering new approaches such as proton beam therapy and immune-related endocrinopathies with the advent of precision oncology treatment. We hope to encourage oncologists and endocrinologists to develop early detection guidelines that minimize sequelae and have a positive impact on their quality of life. Abstract Childhood cancer management has improved considerably over the years, leading to a significant improvement in survival of up to 80%. However, childhood cancer survivors are at the highest risk of developing sequelae resulting from treatment, with endocrine complications being frequently observed among survivors. Multiple predisposing factors for endocrine sequelae have been identified, including age at diagnosis, treatment received, radiation, tumor type, and genetic polymorphisms, which could explain the individual predisposition to develop drug toxicity. Novel agents targeting tumor growth and immune checkpoint inhibitors have recently become the cornerstone for the treatment of different cancers, triggering a myriad of immune-related endocrinopathies. Endocrine sequelae of cancer therapy will have an impact on not only childhood but also on the survival and quality of life of these highly complex patients. Therefore, lifelong monitoring of childhood cancer survivors at risk of endocrine diseases is paramount. Encouraging oncologists and endocrinologists to develop new follow-up and early detection guidelines that minimize sequelae among these patients has become a priority, promoting integration between pediatric and adult units since many sequelae may manifest only after years to decades of follow-up.
Collapse
|
12
|
Kenda M, Avsec D, Zore T, Kogovšek E, Pečar Fonović U, Kos J, Bozovičar K, Bratkovič T, Karas Kuželički N, Žegura B, Filipič M, Sollner Dolenc M. Effects of tyrosine kinase inhibitors on androgen, estrogen α, glucocorticoid and thyroid receptors. Toxicol Appl Pharmacol 2022; 434:115818. [PMID: 34890638 DOI: 10.1016/j.taap.2021.115818] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 11/24/2021] [Accepted: 12/02/2021] [Indexed: 11/25/2022]
Abstract
Modern anticancer therapies favor a targeted approach. Tyrosine kinase inhibitors (TKIs) are drugs that target molecular pathways involved in various types of malignancies. Although TKIs are safe and well tolerated, they remain not completely selective; e.g., endocrine-mediated adverse events have been observed with their use. In the present study, the effects of seven TKIs were determined on the activities of androgen receptor, estrogen receptor α (ERα), glucocorticoid receptor and thyroid receptor in vitro using stably transfected cell lines expressing firefly luciferase reporter gene: AR-EcoScreen, hERα-HeLa9903, MDA-kb2, and GH3.TRE-Luc cells, respectively. Antiandrogenic activity was seen for erlotinib, estrogenic activity for imatinib, antiestrogenic activity for dasatinib, erlotinib, nilotinib, regorafenib and sorafenib, glucocorticoid activity for erlotinib and ibrutinib, antiglucocorticoid activity for regorafenib and sorafenib, and antithyroid activity for ibrutinib. Additionally, synergism was seen for 1-5 μM dasatinib and 500 nM hydrocortisone combination for glucocorticoid activity in MDA-kb2 cells. The estrogenic activity of imatinib was confirmed as mediated through ERα, and interference of the TKIs with the reporter gene assays was ruled out in a cell-lysate-based firefly luciferase enzyme inhibition assay. Imatinib in combination with 4-hydroxytamoxifen showed concentration-dependent effects on the metabolic activity of ERα-expressing AN3CA, MCF-7, and SKOV3 cells, and on cell proliferation and adhesion of MCF-7 cells. These findings contribute to the understanding of the endocrine effects of TKIs, in terms of toxicity and effectiveness, and define the need to further evaluate the endocrine disrupting activities of TKIs to safeguard human and environmental health.
Collapse
Affiliation(s)
- Maša Kenda
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| | - Damjan Avsec
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| | - Taja Zore
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| | - Eva Kogovšek
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000 Ljubljana, Slovenia
| | - Urša Pečar Fonović
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| | - Janko Kos
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| | - Krištof Bozovičar
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| | - Tomaž Bratkovič
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| | | | - Bojana Žegura
- National Institute of Biology, Department of Genetic Toxicology and Cancer Biology, Večna pot 111, SI-1000 Ljubljana, Slovenia.
| | - Metka Filipič
- National Institute of Biology, Department of Genetic Toxicology and Cancer Biology, Večna pot 111, SI-1000 Ljubljana, Slovenia.
| | - Marija Sollner Dolenc
- University of Ljubljana, Faculty of Pharmacy, Aškerčeva 7, SI-1000 Ljubljana, Slovenia.
| |
Collapse
|
13
|
Hayes AG, Rushworth RL, Torpy DJ. Risk assessment, diagnosis, and treatment of cancer treatment-related adrenal insufficiency. Expert Rev Endocrinol Metab 2022; 17:21-33. [PMID: 34979842 DOI: 10.1080/17446651.2022.2023009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/22/2021] [Indexed: 12/17/2022]
Abstract
INTRODUCTION Adrenal insufficiency (AI) is an easily treatable, potentially life-threatening condition, which is increasingly recognized in malignancy. The recent introduction of immune checkpoint inhibitors, in particular, and increasing use of tyrosine kinase inhibitors have increased the frequency of AI in patients with malignancy. A review is therefore warranted to summarize current knowledge on the topic and guide safe clinical practices. AREAS COVERED Malignancy may directly impact the hypothalamic-pituitary-adrenal axis and cause AI, or their treatment including surgery, radiotherapy and medication. In this narrative review, we discuss new causes of AI, recognition of suggestive clinical features, diagnosis and subsequent treatment, aiming to avoid potentially fatal adrenal crisis (AC). Standard literature searching and authors assessment of clinical applicability were used. EXPERT OPINION Adrenal insufficiency can be easily treated once identified but life threatening if unrecognized. While use of new agents such as immune checkpoint inhibitors (ICIs) is increasing, greater understanding of the mechanism of AI is needed to target prediction tools and enhance risk stratification.
Collapse
Affiliation(s)
- Annabelle G Hayes
- Royal Adelaide Hospital, Endocrine and Metabolic Unit Adelaide, Adelaide, Australia
| | - R Louise Rushworth
- The University of Notre Dame Australia, School of Medicine, Sydney Campus Darlinghurst, Darlinghurst, Australia
| | - David J Torpy
- Royal Adelaide Hospital, Endocrine and Metabolic Unit Adelaide, Adelaide, Australia
- University of Adelaide, Discipline of Medicine, Adelaide, Australia
| |
Collapse
|
14
|
Elersek T, Novak M, Mlinar M, Virant I, Bahor N, Leben K, Žegura B, Filipič M. Lethal and Sub-Lethal Effects and Modulation of Gene Expression Induced by T Kinase Inhibitors in Zebrafish (Danio Rerio) Embryos. TOXICS 2021; 10:toxics10010004. [PMID: 35051046 PMCID: PMC8781212 DOI: 10.3390/toxics10010004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 12/16/2021] [Accepted: 12/21/2021] [Indexed: 12/11/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) are designed for targeted cancer therapy. The consumption of these drugs during the last 20 years has been constantly rising. In the zebrafish (Danio rerio) embryo toxicity test, we assessed the toxicity of six TKIs: imatinib mesylate, erlotinib, nilotinib, dasatinib, sorafenib and regorafenib. Imatinib mesylate and dasatinib induced lethal effects, while regorafenib, sorfenib and dasatinib caused a significant increase of sub-lethal effects, predominantly oedema, no blood circulation and formation of blood aggregates. The analyses of the changes in the expression of selected genes associated with the hormone system after the exposure to imatinib mesylate, dasatinib and regorafenib demonstrated that all three tested TKIs deregulated the expression of oestrogen receptor esr1, cytochrome P450 aromatase (cypa19b) and hydroxysteroid-dehydrogenase (hsd3b), regorafenib, and also thyroglobulin (tg). The expression of genes involved in the DNA damage response (gadd45 and mcm6) and apoptosis (bcl2) was deregulated only by exposure to regorafenib. The data indicate that common mechanisms, namely antiangiogenic activity and interference with steroidogenesis are involved in the TKI induced sub-lethal effects and potential hormone disrupting activity, respectively. The residues of TKIs may represent an environmental hazard; therefore, further ecotoxicological studies focusing also on the effects of their mixtures are warranted.
Collapse
Affiliation(s)
- Tina Elersek
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia; (T.E.); (M.N.); (M.M.); (N.B.); (K.L.); (B.Ž.)
| | - Matjaž Novak
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia; (T.E.); (M.N.); (M.M.); (N.B.); (K.L.); (B.Ž.)
| | - Mateja Mlinar
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia; (T.E.); (M.N.); (M.M.); (N.B.); (K.L.); (B.Ž.)
| | - Igor Virant
- Institute of Oncology Ljubljana, Zaloška 2, 1000 Ljubljana, Slovenia;
| | - Nika Bahor
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia; (T.E.); (M.N.); (M.M.); (N.B.); (K.L.); (B.Ž.)
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia
| | - Karin Leben
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia; (T.E.); (M.N.); (M.M.); (N.B.); (K.L.); (B.Ž.)
- Biotechnical Faculty, University of Ljubljana, Jamnikarjeva 101, 1000 Ljubljana, Slovenia
| | - Bojana Žegura
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia; (T.E.); (M.N.); (M.M.); (N.B.); (K.L.); (B.Ž.)
| | - Metka Filipič
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia; (T.E.); (M.N.); (M.M.); (N.B.); (K.L.); (B.Ž.)
- Correspondence:
| |
Collapse
|
15
|
Stress Axis in the Cancer Patient: Clinical Aspects and Management. ENDOCRINES 2021. [DOI: 10.3390/endocrines2040044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Hypothalamus–pituitary–adrenal (HPA) axis alterations are common in cancer patients, mainly due to the different antitumoral therapies, which lead to several acute and late endocrine side effects. This review summarizes the most recent evidence regarding HPA derangement, both in patients with active neoplasms and in cancer survivors, with particular attention to the impact of the different antitumoral treatments, focusing on the major clinical aspects. While acute hormone failure usually results from injury caused directly by tumor burden or surgical interventions, short- and long-term effects are generally due to chemotherapy, radiotherapy and, as more recently shown, to different types of targeted- and immuno-therapy. Adrenal insufficiency (AI) is mostly caused by pituitary or hypothalamic injury rather than a direct damage of the adrenal gland. Moreover, other treatments commonly employed as supportive therapy or in the context of palliative care (i.e., glucocorticoids, opioids) can lead to HPA dysfunction. Epidemiology and pathophysiology of stress axis alterations in cancer patients still require clarification. Since AI may represent a life-threatening condition, monitoring adrenal function in cancer patients is mandatory, especially in subjects who experience fatigue or during stress conditions, in order to promptly start replacement treatment when needed.
Collapse
|
16
|
Lorenzi E, Simonelli M, Persico P, Dipasquale A, Santoro A. Risks of molecular targeted therapies to fertility and safety during pregnancy: a review of current knowledge and future needs. Expert Opin Drug Saf 2021; 20:503-521. [PMID: 33600273 DOI: 10.1080/14740338.2021.1893299] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION As the population of young cancer survivors is increasing and a trend toward postponing pregnancy later in life is reported, more efforts are focused toward understanding treatment-induced sequelae, in particular, the effects of cancer and/or treatment on fertility. AREA COVERED Whereas the fertility risk of cytotoxic agents for both men and women is well recognized, the impact of molecular-targeted therapy (MTT) on fertility parameters, their teratogenic potential and pregnancy outcome/management in case of an accidental exposure are not established. We update available clinical data on the impact of new MTTs on fertility in both sexes, their potential teratogenic effects and the outcome of pregnancy during accidental exposure. Agents are categorized by class and the potential relevance of their target signaling pathways to gonadal maturation. EXPERT OPINION The majority of MTTs have worrying preclinical data discouraging their use during pregnancy and reinforcing the idea that they can induce impairment in gonadal function. However, it does not mean that all MTTs result in permanent infertility and that they should be completely avoided during pregnancy. The current review provides a critical evaluation on the most commonly used MTTs, offering a possible guide for clinicians.
Collapse
Affiliation(s)
- Elena Lorenzi
- Department of Oncology, IRCCS Humanitas Cancer Center, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Pieve Emanuele, Italy
| | - Matteo Simonelli
- Department of Oncology, IRCCS Humanitas Cancer Center, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Pieve Emanuele, Italy
| | - Pasquale Persico
- Department of Oncology, IRCCS Humanitas Cancer Center, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Pieve Emanuele, Italy
| | - Angelo Dipasquale
- Department of Oncology, IRCCS Humanitas Cancer Center, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Pieve Emanuele, Italy
| | - Armando Santoro
- Department of Oncology, IRCCS Humanitas Cancer Center, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Pieve Emanuele, Italy
| |
Collapse
|
17
|
Novak M, Baebler Š, Žegura B, Rotter A, Gajski G, Gerić M, Garaj-Vrhovac V, Bakos K, Csenki Z, Kovács R, Horváth Á, Gazsi G, Filipič M. Deregulation of whole-transcriptome gene expression in zebrafish (Danio rerio) after chronic exposure to low doses of imatinib mesylate in a complete life cycle study. CHEMOSPHERE 2021; 263:128097. [PMID: 33297093 DOI: 10.1016/j.chemosphere.2020.128097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 06/12/2023]
Abstract
Imatinib mesylate (IM) is an anticancer drug that belongs to tyrosine kinase inhibitors. We report the results of the first investigation of the chronic exposure of zebrafish (Danio rerio) to IM. The exposure to IM (0.01, 1 and 100 μg/L) was initiated in adult fish and continued through hatching and the offspring generation for seven months. In addition to standard toxicological endpoints, induction of genotoxic effects and whole-genome transcriptome of liver samples of offspring generation of zebrafish were analysed. Exposure to IM did not affect the survival and growth of zebrafish, did not cause any histopathological changes, but it induced a marginal increase in the chromosomal damage in blood cells. The whole-genome transcriptome analyses demonstrated dose-dependent increase in the number of differentially expressed genes with a significantly higher number of deregulated genes in female fish compared to male. Differentially expressed genes included genes involved in response to DNA damage, cell cycle control and regulation of circadian rhythm. Based on the low genotoxic activity and the pattern of the changes in DNA damage responsive genes we consider that at current environmental exposure levels, IM represents low risk for genotoxic effects in aquatic organisms. Exposure to IM also induced deregulation of the expression of genes associated with steroidogenesis and hormone metabolism and function, which indicates hormone-disrupting activity of IM that has not been studied so far. The study provide new information on the potential consequences of chronic exposure to the residues of tyrosine kinase inhibitors, which remain to be further explored.
Collapse
Affiliation(s)
- Matjaž Novak
- National Institute of Biology, Večna Pot 111, 1000, Ljubljana, Slovenia.
| | - Špela Baebler
- National Institute of Biology, Večna Pot 111, 1000, Ljubljana, Slovenia
| | - Bojana Žegura
- National Institute of Biology, Večna Pot 111, 1000, Ljubljana, Slovenia
| | - Ana Rotter
- National Institute of Biology, Večna Pot 111, 1000, Ljubljana, Slovenia
| | - Goran Gajski
- Institute for Medical Research and Occupational Health, Mutagenesis Unit, Ksaverska Cesta 2, 10000, Zagreb, Croatia
| | - Marko Gerić
- Institute for Medical Research and Occupational Health, Mutagenesis Unit, Ksaverska Cesta 2, 10000, Zagreb, Croatia
| | - Vera Garaj-Vrhovac
- Institute for Medical Research and Occupational Health, Mutagenesis Unit, Ksaverska Cesta 2, 10000, Zagreb, Croatia
| | - Katalin Bakos
- Department of Aquaculture, Institute for Conservation of Natural Resources, Faculty of Agricultural and Environmental Sciences, Szent István University, 1. Pater Károly U, H-2100, Gödöllo, Hungary
| | - Zsolt Csenki
- Department of Aquaculture, Institute for Conservation of Natural Resources, Faculty of Agricultural and Environmental Sciences, Szent István University, 1. Pater Károly U, H-2100, Gödöllo, Hungary
| | - Róbert Kovács
- Department of Aquaculture, Institute for Conservation of Natural Resources, Faculty of Agricultural and Environmental Sciences, Szent István University, 1. Pater Károly U, H-2100, Gödöllo, Hungary
| | - Ákos Horváth
- Department of Aquaculture, Institute for Conservation of Natural Resources, Faculty of Agricultural and Environmental Sciences, Szent István University, 1. Pater Károly U, H-2100, Gödöllo, Hungary
| | - Gyöngyi Gazsi
- Department of Aquaculture, Institute for Conservation of Natural Resources, Faculty of Agricultural and Environmental Sciences, Szent István University, 1. Pater Károly U, H-2100, Gödöllo, Hungary
| | - Metka Filipič
- National Institute of Biology, Večna Pot 111, 1000, Ljubljana, Slovenia
| |
Collapse
|
18
|
van Santen HM, Chemaitilly W, Meacham LR, Tonorezos ES, Mostoufi-Moab S. Endocrine Health in Childhood Cancer Survivors. Pediatr Clin North Am 2020; 67:1171-1186. [PMID: 33131540 DOI: 10.1016/j.pcl.2020.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Endocrine late effects, including reproductive disorders and secondary thyroid cancer, have been reported in up to 50 %childhood cancer survivors (CCS) more than 5 years after treatment. Most endocrine disorders are amenable to treatment; awareness of symptoms is therefore of great importance. Recognition of these symptoms may be delayed however because many are nonspecific. Timely treatment of endocrine disorders improves quality of life in CCS and prevents possible consequences, such as short stature, bone and cardiovascular disorders, and depression. At-risk CCS must therefore be regularly and systematically monitored. This article provides a summary of the most commonly reported endocrine late effects in CCS.
Collapse
Affiliation(s)
- Hanneke M van Santen
- Department of Pediatric Endocrinology, Wilhelmina Children's Hospital, UMCU, PO Box 85090, Utrecht 3505 AB, the Netherlands; Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| | - Wassim Chemaitilly
- Division of Endocrinology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Lillian R Meacham
- Division of Hematology/Oncology/BMT, Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Emily S Tonorezos
- Department of Medicine, Memorial Sloan Kettering and Weill Cornell Medical College, Memorial Sloan Kettering Cancer Center, 485 Lexington Avenue, 2nd Floor, New York, NY 10017, USA
| | - Sogol Mostoufi-Moab
- Division of Endocrinology, Department of Pediatrics, The Children's Hospital of Philadelphia, Roberts Pediatric Clinical Research Building, 2716 South Street, Philadelphia, PA 19146, USA; Division of Oncology, Department of Pediatrics, The Children's Hospital of Philadelphia, Roberts Pediatric Clinical Research Building, 2716 South Street, Philadelphia, PA 19146, USA
| |
Collapse
|
19
|
Qiu X, Li M, Wu L, Xin Y, Mu S, Li T, Song K. Severe Fatigue is an Important Factor in the Prognosis of Patients with Advanced Hepatocellular Carcinoma Treated with Sorafenib. Cancer Manag Res 2020; 12:7983-7992. [PMID: 32943932 PMCID: PMC7481349 DOI: 10.2147/cmar.s233448] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 08/03/2020] [Indexed: 12/11/2022] Open
Abstract
Objective To investigate the effects of fatigue on the survival of patients with advanced hepatocellular carcinoma treated with sorafenib. Patients and Methods A retrospective analysis of 182 cases of patients with advanced hepatocellular carcinoma treated with sorafenib in our hospital from October 1, 2008, to October 31, 2017, showed clinical and pathological data and follow-up results. The clinical and pathological data as well as follow-up results of 182 patients with advanced hepatocellular carcinoma treated with sorafenib in our hospital from October 1, 2008, to October 31, 2018, were retrospectively analyzed. All patients were treated for at least 3 months. Patients were divided into three groups: fatigue grade I (n=74), fatigue grade II (n=62), and fatigue grade III (n=46), according to National Cancer Institute common terminology criteria for adverse events (NCI CTCAE) version 5.0. Survival analysis between groups was performed by the Kaplan–Meier method (Log rank test), continuous variables were analyzed by t-test, and categorical variables were analyzed by chi-square test. Results The overall survival (OS) of patients who were relieved of fatigue was 33.0±9.3 months, whereas the OS of patients who were not relieved of fatigue was 15.0±1.8 months (P<0.000). Furthermore, the time to progress (TTP) of patients who were relieved of fatigue by resting was 20.3 ± 10.9 months compared to a TTP of 7.7 ± 1.0 months in patients who were not relieved of fatigue (P<0.000). Conclusion Patients, especially the elderly and infirm, were more susceptible to toxicity.
Collapse
Affiliation(s)
- Xuan Qiu
- Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Manjiang Li
- Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Liqun Wu
- Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Yang Xin
- Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Siyu Mu
- Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Tianxiang Li
- Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| | - Kangjian Song
- Liver Disease Center, The Affiliated Hospital of Qingdao University, Qingdao 266003, People's Republic of China
| |
Collapse
|
20
|
Derry PJ, Hegde ML, Jackson GR, Kayed R, Tour JM, Tsai AL, Kent TA. Revisiting the intersection of amyloid, pathologically modified tau and iron in Alzheimer's disease from a ferroptosis perspective. Prog Neurobiol 2020; 184:101716. [PMID: 31604111 PMCID: PMC7850812 DOI: 10.1016/j.pneurobio.2019.101716] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 08/12/2019] [Accepted: 09/26/2019] [Indexed: 02/06/2023]
Abstract
The complexity of Alzheimer's disease (AD) complicates the search for effective treatments. While the key roles of pathologically modified proteins has occupied a central role in hypotheses of the pathophysiology, less attention has been paid to the potential role for transition metals overload, subsequent oxidative stress, and tissue injury. The association of transition metals, the major focus heretofore iron and amyloid, the same can now be said for the likely pathogenic microtubular associated tau (MAPT). This review discusses the interplay between iron, pathologically modified tau and oxidative stress, and connects many related discoveries. Basic principles of the transition to pathological MAPT are discussed. Iron, its homeostatic mechanisms, the recently described phenomenon of ferroptosis and purported, although still controversial roles in AD are reviewed as well as considerations to overcome existing hurdles of iron-targeted therapeutic avenues that have been attempted in AD. We summarize the involvement of multiple pathological pathways at different disease stages of disease progression that supports the potential for a combinatorial treatment strategy targeting multiple factors.
Collapse
Affiliation(s)
- Paul J Derry
- Center for Genomics and Precision Medicine, Institute of Biosciences and Technology, College of Medicine, Texas A&M Health Science Center, Houston, TX, United States
| | - Muralidhar L Hegde
- Institute for Academic Medicine, Houston Methodist, Weill Cornell Medical College, Houston, TX, United States
| | - George R Jackson
- Department of Neurology Baylor College of Medicine, Houston, TX, United States; Parkinson's Disease Research, Education and Clinical Center (PADRECC), Michael E. DeBakey VA Medical Center, Houston, TX, United States
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Disorders, Department of Neurology, University of Texas Medical Branch, Galveston, TX, United States
| | - James M Tour
- Smalley Institute for Nanoscale Science and Technology, Rice University, Houston, TX, United States
| | - Ah-Lim Tsai
- Department of Biochemistry and Hematology, McGovern School of Medicine, UT Health Science Center, Houston, TX, United States
| | - Thomas A Kent
- Center for Genomics and Precision Medicine, Institute of Biosciences and Technology, College of Medicine, Texas A&M Health Science Center, Houston, TX, United States; Department of Chemistry, Rice University, Houston, TX, United States; Stanley H. Appel Department of Neurology, Houston Methodist Hospital, Houston, TX, United States.
| |
Collapse
|
21
|
Imatinib mesylate effects on zebrafish reproductive success: Gonadal development, gamete quality, fertility, embryo-larvae viability and development, and related genes. Toxicol Appl Pharmacol 2019; 379:114645. [PMID: 31278918 DOI: 10.1016/j.taap.2019.114645] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 05/24/2019] [Accepted: 07/01/2019] [Indexed: 11/20/2022]
Abstract
Imatinib (IM) is a tyrosine kinase (TK) inhibitor (TKI) used to treat chronic myeloid leukemia. Clinical case reports and a few laboratory mammal studies provide inconclusive evidence about its deleterious effects on reproduction. The aim of the current study was to evaluate the potential of zebrafish to characterize IM-induced effects on reproduction and clarify IM effects on reproductive success. To this end, we exposed adult zebrafish to four concentrations of IM for 30 days followed by a 30-day depuration period. IM exposure caused a concentration-dependent, irreversible, suppression of folliculogenesis, reversible decrease in sperm density and motility, decreased fecundity and fertility, but no significant change in atretic follicle abundance. We also observed IM-induced premature hatching, but no significant change in embryo-larvae survivability. However, we found significant IM-induced morphometric malformations. IM decreased expression of vegfaa and igf2a (two reproductive-, angiogenic-, and growth-related genes) in testes and ovaries. The results demonstrate IM can induce significant changes in critical reproductive endpoints and zebrafish as a suitable model organism to show effects of IM on reproduction. The findings suggest that TKI effects on reproductive success should be considered.
Collapse
|
22
|
Li J, Halfter K, Zhang M, Saad C, Xu K, Bauer B, Huang Y, Shi L, Mansmann UR. Computational analysis of receptor tyrosine kinase inhibitors and cancer metabolism: implications for treatment and discovery of potential therapeutic signatures. BMC Cancer 2019; 19:600. [PMID: 31208363 PMCID: PMC6580552 DOI: 10.1186/s12885-019-5804-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Accepted: 06/06/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Receptor tyrosine kinase (RTK) inhibitors are frequently used to treat cancers and the results have been mixed, some of these small molecule drugs are highly successful while others show a more modest response. A high number of studies have been conducted to investigate the signaling mechanisms and corresponding therapeutic influence of RTK inhibitors in order to explore the therapeutic potential of RTK inhibitors. However, most of these studies neglected the potential metabolic impact of RTK inhibitors, which could be highly associated with drug efficacy and adverse effects during treatment. METHODS In order to fill these knowledge gaps and improve the therapeutic utilization of RTK inhibitors a large-scale computational simulation/analysis over multiple types of cancers with the treatment responses of RTK inhibitors was performed. The pharmacological data of all eight RTK inhibitor and gene expression profiles of 479 cell lines from The Cancer Cell Line Encyclopedia were used. RESULTS The potential metabolic impact of RTK inhibitors on different types of cancers were analyzed resulting in cancer-specific (breast, liver, pancreas, central nervous system) metabolic signatures. Many of these are in line with results from different independent studies, thereby providing indirect verification of the obtained results. CONCLUSIONS Our study demonstrates the potential of using a computational approach on signature-based-analysis over multiple cancer types. The results reveal the strength of multiple-cancer analysis over conventional signature-based analysis on a single cancer type.
Collapse
Affiliation(s)
- Jian Li
- Institute for Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-University München, Munich, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Kathrin Halfter
- Institute for Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-University München, Munich, Germany
| | - Mengying Zhang
- Institute for Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-University München, Munich, Germany
| | - Christian Saad
- Department of Computational Science, University of Augsburg, Augsburg, Germany
| | - Kai Xu
- Department of Orthopaedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Bernhard Bauer
- Department of Computational Science, University of Augsburg, Augsburg, Germany
| | - Yijiang Huang
- Department of Orthopaedics, Physical Medicine and Rehabilitation, University Hospital, LMU, Munich, Germany
| | - Lei Shi
- Institute of Photomedicine, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Ulrich R. Mansmann
- Institute for Medical Informatics, Biometry and Epidemiology, Ludwig-Maximilians-University München, Munich, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
23
|
Afshar M, Patel HRH, Jain A, Kumar A, Patel P, James ND, Porfiri E. Chronic tyrosine kinase inhibitor (TKI) use in metastatic renal cell carcinoma (mRCC): can this lead to the adverse effect of hypogonadism? Expert Rev Anticancer Ther 2019; 19:529-532. [PMID: 30995130 DOI: 10.1080/14737140.2019.1609355] [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: 12/21/2018] [Accepted: 04/16/2019] [Indexed: 10/27/2022]
Abstract
Background: Patients with metastatic renal cell carcinoma (mRCC) are commonly treated with tyrosine kinase inhibitors (TKIs). An adverse effect frequently suffered by patients is lethargy, which often leads to dose reduction or drug cessation. We aimed to assess whether hypogonadism is related to treatment with TKIs. Methods: We prospectively assessed gonadal function in 41 consecutive males with mRCC treated with TKIs. Demographic, clinical, and biochemical variables were collected, and statistical analyses performed to assess correlation and survival. Data Capture for each patient was perfomred at the time of entry in the study. Results: There was a 77% incidence of hypogonadism in this cohort. Assessment of testosterone level and time on TKI treatment revealed a correlation with linear regression R2 of 0.24 and regression coefficient of -0.003 (p = 0.019). Odds ratio for hypogonadism at >30 months on TKIs was 12.1 (p = 0.011). Odds ratios above and below this value showed a confirmatory trend, suggesting that this may be a chronic adverse effect. Conclusions: Our findings provide an important and robust hypothesis for a prospective clinical trial to be performed. Expert Opinion: Given the present data, patients who have symptoms suggestive of hypogonadism must have an assessment of gonadal function and be treated.
Collapse
Affiliation(s)
- Mehran Afshar
- a Department of Oncology , St George's University Hospital NHS Foundation Trust , London , UK
| | - Hiten R H Patel
- b Department of Urology , University of Tromso , Tromso , Norway
| | - Ankit Jain
- c Department of Oncology , The Royal Wolverhampton NHS Trust , Wolverhampton , UK
| | - Abhishek Kumar
- d Department of Statistics , Indira Gandhi Institute of Development Research , Mumbai , India
| | - Prashant Patel
- e Institute of Cancer and Genomic Sciences , University of Birmingham , Birmingham , UK
| | - Nicholas D James
- e Institute of Cancer and Genomic Sciences , University of Birmingham , Birmingham , UK
- f The Cancer Centre , Queen Elizabeth Hospital Birmingham , Birmingham , UK
| | - Emilio Porfiri
- f The Cancer Centre , Queen Elizabeth Hospital Birmingham , Birmingham , UK
| |
Collapse
|
24
|
Sklar CA, Antal Z, Chemaitilly W, Cohen LE, Follin C, Meacham LR, Murad MH. Hypothalamic-Pituitary and Growth Disorders in Survivors of Childhood Cancer: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab 2018; 103:2761-2784. [PMID: 29982476 DOI: 10.1210/jc.2018-01175] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 05/29/2018] [Indexed: 11/19/2022]
Abstract
OBJECTIVE To formulate clinical practice guidelines for the endocrine treatment of hypothalamic-pituitary and growth disorders in survivors of childhood cancer. PARTICIPANTS An Endocrine Society-appointed guideline writing committee of six medical experts and a methodologist. CONCLUSIONS Due to remarkable improvements in childhood cancer treatment and supportive care during the past several decades, 5-year survival rates for childhood cancer currently are >80%. However, by virtue of their disease and its treatments, childhood cancer survivors are at increased risk for a wide range of serious health conditions, including disorders of the endocrine system. Recent data indicate that 40% to 50% of survivors will develop an endocrine disorder during their lifetime. Risk factors for endocrine complications include both host (e.g., age, sex) and treatment factors (e.g., radiation). Radiation exposure to key endocrine organs (e.g., hypothalamus, pituitary, thyroid, and gonads) places cancer survivors at the highest risk of developing an endocrine abnormality over time; these endocrinopathies can develop decades following cancer treatment, underscoring the importance of lifelong surveillance. The following guideline addresses the diagnosis and treatment of hypothalamic-pituitary and growth disorders commonly encountered in childhood cancer survivors.
Collapse
Affiliation(s)
| | - Zoltan Antal
- Memorial Sloan-Kettering Cancer Center, New York, New York
- Weill Cornell Medicine and New York Presbyterian Hospital, New York, New York
| | | | | | | | | | - M Hassan Murad
- Mayo Clinic Evidence-Based Practice Center, Rochester, Minnesota
| |
Collapse
|
25
|
Koyama S, Miyake N, Fujiwara K, Morisaki T, Fukuhara T, Kitano H, Takeuchi H. Lenvatinib for Anaplastic Thyroid Cancer and Lenvatinib-Induced Thyroid Dysfunction. Eur Thyroid J 2018; 7:139-144. [PMID: 30023346 PMCID: PMC6047489 DOI: 10.1159/000485972] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/01/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Lenvatinib is an oral multitargeted tyrosine kinase inhibitor that has an anticancer action in patients with differentiated thyroid cancer that is refractory to radioiodine. Knowledge of the efficacy and safety of lenvatinib in patients with anaplastic thyroid cancer (ATC) is limited. Tyrosine kinase inhibitors frequently cause hypothyroidism, but the incidence of hypothyroidism with lenvatinib is unclear. OBJECTIVES We conducted a retrospective study to investigate the efficacy and safety of lenvatinib in ATC. METHODS Five patients with unresectable ATC were enrolled. Lenvatinib 24 mg once daily was administered until disease progression, unmanageable toxicity, withdrawal, or death occurred. We retrospectively analyzed the objective response rate (ORR), time to progression (TTP), overall survival, and safety. RESULTS Three of the 5 patients (60%) had a partial response, and 5 (40%) had stable disease. The ORR was 60%. Median TTP was 88 days, and overall survival was 165 days. Hypothyroidism was a common treatment-related adverse effect; 4 patients (80%) had hypothyroidism of any grade. These 4 patients had not undergone total thyroidectomy prior to lenvatinib administration, and the other patient had undergone total thyroidectomy. Treatment-related adverse effects of any grade were hypertension in 80% of patients, diarrhea in 40%, fatigue in 80%, and decreased appetite in 80%. CONCLUSIONS Lenvatinib is an effective treatment and may improve the prognosis of unresectable ATC. Four of the 5 patients had hypothyroidism, which may have been associated with treatment-induced injury of the thyroid gland. There were many treatment-related adverse effects, most of which were manageable by dose modification and medical therapy.
Collapse
Affiliation(s)
- Satoshi Koyama
- Department of Otolaryngology Head and Neck Surgery, Tottori University Faculty of Medicine, Yonago, Japan
- *Satoshi Koyama, MD, Department of Otolaryngology Head and Neck Surgery, Tottori University Faculty of Medicine, 36-1 Nishicho, Yonago, Tottori 683-8504 (Japan), E-Mail
| | - Naritomo Miyake
- Department of Otolaryngology Head and Neck Surgery, Tottori University Faculty of Medicine, Yonago, Japan
| | - Kazunori Fujiwara
- Department of Otolaryngology Head and Neck Surgery, Tottori University Faculty of Medicine, Yonago, Japan
| | - Tsuyoshi Morisaki
- Department of Otolaryngology Head and Neck Surgery, Tottori University Faculty of Medicine, Yonago, Japan
| | - Takahiro Fukuhara
- Department of Otolaryngology Head and Neck Surgery, Tottori University Faculty of Medicine, Yonago, Japan
| | - Hiroya Kitano
- Department of Otolaryngology Head and Neck Surgery, Tottori University Faculty of Medicine, Yonago, Japan
- Center for Head and Neck Surgery, Kusatsu General Hospital, Kusatsu, Japan
| | - Hiromi Takeuchi
- Department of Otolaryngology Head and Neck Surgery, Tottori University Faculty of Medicine, Yonago, Japan
| |
Collapse
|
26
|
Patel NS, Oury A, Daniels GA, Bazhenova L, Patel SP. Incidence of Thyroid Function Test Abnormalities in Patients Receiving Immune-Checkpoint Inhibitors for Cancer Treatment. Oncologist 2018; 23:1236-1241. [PMID: 29769383 DOI: 10.1634/theoncologist.2017-0375] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 03/14/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND With the advent of immune-checkpoint inhibitor (ICI) therapy (anti-CTLA-4, anti-PD-1), immune-related adverse events such as thyroid function test abnormalities (TFTAs) are common, with a reported incidence range of 2%-15% depending upon the ICI used. The aim of this study is to describe the incidence of TFTAs retrospectively in patients who received ICI therapy. METHODS A total of 285 patients were reviewed (178 male, 107 female; 16-94 years of age), of whom 218 had no baseline TFTAs, 61 had baseline TFTAs, and 6 had a history of thyroidectomy (excluded). At least one dose of ipilimumab and/or nivolumab or pembrolizumab was administered. Post-ICI therapy TFTAs were classified according to standard definitions of thyroid conditions when possible. RESULTS A total of 35% (76/218) patients had new-onset TFTAs on ICI therapy. Of note, 70.5% (43/61) had baseline TFTAs that were exacerbated by ICI therapy. The median times to new-onset or exacerbated baseline TFTA were 46 and 33 days, respectively. Of note, 64.5% (20/31) of patients on both ipilimumab and nivolumab had new-onset TFTAs, compared with 31.3% (15/48) on ipilimumab, 31.5% (28/89) on nivolumab, and 26% (13/50) on pembrolizumab. CONCLUSION The incidence of TFTAs with ICI therapy was higher than previously reported. Patients with baseline TFTAs and/or who were receiving ipilimumab and nivolumab combination therapy had a higher incidence of TFTAs than patients receiving single-agent ICI therapy. We recommend more frequent evaluation of thyroid function in the first 8 weeks, especially in patients with baseline TFTAs. IMPLICATIONS FOR PRACTICE Increased use of immune-checkpoint inhibitors in cancer treatment has highlighted the importance of monitoring for and treating immune-related adverse events. This study was conducted to assess the incidence of thyroid function test abnormalities retrospectively in patients with cancer on immune-checkpoint inhibitors, which is not known exactly. This study is unique in that it included patients with a variety of histologic subtypes of cancer and also followed the clinical course of patients with baseline thyroid function test abnormalities. This study can help make oncologists aware that the incidence of thyroid function test abnormalities is higher than anticipated. Early identification and timely treatment can help ameliorate symptoms for patients and improve their overall quality of life.
Collapse
Affiliation(s)
- Nisha Subhash Patel
- Department of Hematology & Oncology, University of California San Diego, Moores Cancer Center, San Diego, California, USA
| | - Anais Oury
- Department of Hematology & Oncology, University of California San Diego, Moores Cancer Center, San Diego, California, USA
| | - Gregory A Daniels
- Department of Hematology & Oncology, University of California San Diego, Moores Cancer Center, San Diego, California, USA
| | - Lyudmila Bazhenova
- Department of Hematology & Oncology, University of California San Diego, Moores Cancer Center, San Diego, California, USA
| | - Sandip Pravin Patel
- Department of Hematology & Oncology, University of California San Diego, Moores Cancer Center, San Diego, California, USA
| |
Collapse
|
27
|
Yazdi MH, Faramarzi MA, Nikfar S, Abdollahi M. Comparative safety and efficacy of tyrosine kinase inhibitors (TKIs) in the treatment setting of different types of leukemia, and different types of adenocarcinoma. Biomed Pharmacother 2017; 95:1556-1564. [PMID: 28950655 DOI: 10.1016/j.biopha.2017.09.088] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 09/17/2017] [Accepted: 09/18/2017] [Indexed: 02/01/2023] Open
Abstract
Tyrosine kinase inhibitors (TKIs) are blockers of tyrosine kinase proteins which are known for the activation of signaling pathways especially in neoplastic cells. TKIs are among targeted anticancer medications that are available in the market. Imatinib was introduced in the late 1990s as the first TKI medicine in oncology, followed by gefitinib, erlotinib, sorafenib, sunitinib, dasatanib and the list of TKIs is being updated nearly every month. To review the safety, efficacy, and current clinical stage of TKIs in different malignancies, particularly leukemia, advanced gastrointestinal and breast cancer, whole literature over the last decade (2006 to 2017) were searched to find all related studies. Criticizing current data indicates that TKIs have shown better clinical outcome in terms of both safety and efficacy compared to conventional therapies. Meanwhile, regarding the results of available clinical trials, the best approach into maximizing the benefits of this novel targeting therapy and also minimizing the undesirable adverse effects, is to evaluate the pharmacogenetic data of patients before allocating these agents in their treatment setting.
Collapse
Affiliation(s)
- Mohammad Hossein Yazdi
- Department of Pharmaceutical Biotechnology and Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Evidence-Based Medicine Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology and Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Shekoufeh Nikfar
- Evidence-Based Medicine Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdollahi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Toxicology and Diseases Group, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Wang X, Yao J, Wang J, Zhang Q, Brady SW, Arun B, Seewaldt VL, Yu D. Targeting Aberrant p70S6K Activation for Estrogen Receptor-Negative Breast Cancer Prevention. Cancer Prev Res (Phila) 2017; 10:641-650. [PMID: 28877935 DOI: 10.1158/1940-6207.capr-17-0106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 07/12/2017] [Accepted: 08/28/2017] [Indexed: 12/27/2022]
Abstract
The prevention of estrogen receptor-negative (ER-) breast cancer remains a major challenge in the cancer prevention field, although antiestrogen and aromatase inhibitors have shown adequate efficacy in preventing estrogen receptor-positive (ER+) breast cancer. Lack of commonly expressed, druggable targets is a major obstacle for meeting this challenge. Previously, we detected the activation of Akt signaling pathway in atypical hyperplasic early-stage lesions of patients. In the current study, we found that Akt and the downstream 70 kDa ribosomal protein S6 kinase (p70S6K) signaling pathway was highly activated in ER- premalignant breast lesions and ER- breast cancer. In addition, p70S6K activation induced transformation of ER- human mammary epithelial cells (hMEC). Therefore, we explored the potential of targeting Akt/p70S6K in the p70S6K activated, ER- hMEC models and mouse mammary tumor models for the prevention of ER- breast cancer. We found that a clinically applicable Akt/p70S6K dual inhibitor, LY2780301, drastically decreased proliferation of hMECs with ErbB2-induced p70S6K activation via Cyclin B1 inhibition and cell-cycle blockade at G0-G1 phase, while it did not significantly reverse the abnormal acinar morphology of these hMECs. In addition, a brief treatment of LY2780301 in MMTV-neu mice that developed atypical hyperplasia (ADH) and mammary intraepithelial neoplasia (MIN) lesions with activated p70S6K was sufficient to suppress S6 phosphorylation and decrease cell proliferation in hyperplasic MECs. In summary, targeting the aberrant Akt/p70S6K activation in ER- hMEC models in vitro and in the MMTV-neu transgenic mouse model in vivo effectively inhibited Akt/S6K signaling and reduced proliferation of hMECs in vitro and ADH/MIN lesions in vivo, indicating its potential in prevention of p70S6K activated ER- breast cancer. Cancer Prev Res; 10(11); 641-50. ©2017 AACR.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jun Yao
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jinyang Wang
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Qingling Zhang
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Samuel W Brady
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas.,Cancer Biology Program, University of Texas Graduate School of Biomedical Sciences - Houston, Houston, Texas
| | - Banu Arun
- Department of Breast Medical Oncology and Clinical Cancer Genetics Program, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Victoria L Seewaldt
- Department of Population Sciences, City of Hope, Duarte, California.,Comprehensive Cancer Center, City of Hope, Duarte, California
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas. .,Cancer Biology Program, University of Texas Graduate School of Biomedical Sciences - Houston, Houston, Texas.,Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
29
|
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
|
30
|
Rzechorzek NM, Liuti T, Stalin C, Marioni-Henry K. Restored vision in a young dog following corticosteroid treatment of presumptive hypophysitis. BMC Vet Res 2017; 13:63. [PMID: 28241874 PMCID: PMC5330113 DOI: 10.1186/s12917-017-0983-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2017] [Accepted: 02/21/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hypophysitis is an umbrella term for a group of disorders involving inflammation of the pituitary gland. A rare occurrence in humans, hypophysitis can produce a range of clinical signs including (but not limited to) visual deficits and diabetes insipidus. Only five cases of canine hypophysitis exist in the literature, all presenting in mature dogs with no visual deficits and a grave outcome. This case report describes the clinical and advanced imaging features of blindness-inducing presumptive hypophysitis in a dog, which rapidly resolved with medical management. CASE PRESENTATION A 1-year-and-seven-month-old neutered male Standard Poodle presented with subacute blindness, ataxia, and polyuria/polydipsia (PUPD). Magnetic resonance imaging (MRI) detected a contrast-enhancing pituitary mass with perilesional oedema compromising the optic chiasm. Suspecting neoplasia, anti-inflammatory corticosteroid was commenced prior to radiation therapy planning. Complete resolution of neurological and visual deficits occurred within 12 days of starting steroid treatment. Repeated advanced imaging indicated macroscopic resolution of the lesion. An extended thyroid panel with insulin-like growth factor-1 analysis supported a diagnosis of hypophysitis. Resolution of PUPD was achieved with tapering courses of prednisolone and desmopressin; the dog has since been clinically normal for 14 months and treatment-free for 11 months. CONCLUSIONS To the authors' knowledge, this is the first instance in which a canine pituitary mass has demonstrated long-term resolution with palliative medical treatment alone, alongside reversal of associated blindness and presumptive diabetes insipidus. We suspect this lesion to be a form of hypophysitis, which should be included among differential diagnoses for pituitary masses, and for subacute blindness in dogs. Where possible, we advocate biopsy-confirmation of hypophysitis prior to timely intervention with anti-inflammatory treatment.
Collapse
Affiliation(s)
- Nina Marie Rzechorzek
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB UK
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush Campus, Roslin, Midlothian EH25 9RG UK
| | - Tiziana Liuti
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush Campus, Roslin, Midlothian EH25 9RG UK
| | - Catherine Stalin
- The Neurology Service, Small Animal Hospital, School of Veterinary Medicine, University of Glasgow, Garscube Campus, Bearsden Road, Glasgow, G61 1QH UK
| | - Katia Marioni-Henry
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush Campus, Roslin, Midlothian EH25 9RG UK
| |
Collapse
|
31
|
Transient hypophysitis in a patient treated by duvelisib for chronic lymphocytic leukemia. Ann Hematol 2017; 96:685-686. [DOI: 10.1007/s00277-017-2928-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 01/17/2017] [Indexed: 11/25/2022]
|
32
|
Labochka D, Moszczuk B, Kukwa W, Szczylik C, Czarnecka AM. Mechanisms through which diabetes mellitus influences renal cell carcinoma development and treatment: A review of the literature. Int J Mol Med 2016; 38:1887-1894. [PMID: 27748835 DOI: 10.3892/ijmm.2016.2776] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 10/06/2016] [Indexed: 11/05/2022] Open
|
33
|
Gallo M, Gentile L, Arvat E, Bertetto O, Clemente G. Diabetology and oncology meet in a network model: union is strength. Acta Diabetol 2016; 53:515-24. [PMID: 26862082 DOI: 10.1007/s00592-016-0839-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 01/10/2016] [Indexed: 12/13/2022]
Abstract
Diabetes and cancer are increasingly common conditions, and the management of cancer patients with diabetes is often challenging. Diabetes in cancer patients poses several complex clinical issues, including which treatment is suitable to control hyperglycemia, how to better counteract glucocorticoid-induced hyperglycemia, and how to manage nutritional problems of cachectic patients and glucose variability linked to artificial nutrition. A key aspect to consider is the patients' position on the trajectory of the oncologic disease, both to establish which level of glycemic control should be pursued and to decide the most suitable antidiabetic treatment to recommend. Endocrinologists are rarely involved in the management of patients with advanced cancer. Furthermore, lack of guidelines results in a "trial-and-error" approach, often with suboptimal disease management. Lastly, cancer survivors represent a frequently underestimated category of patients at higher cardiometabolic risk. A practical solution for these challenges lies in the implementation of care networks based on a close partnership and ongoing communication between oncologists, endocrinologists, and nutritionists, placing the patient at the center of the care process. At the same time, universities and scientific societies should play a key role in promoting research into areas of intersection of oncology and endocrinology, in raising awareness of common possibilities of primary and secondary prevention of metabolic and oncologic diseases, as well as specific challenges of managing diabetes and cancer, and proper training of health workers, while also supporting the shared implementation of effective management strategies.
Collapse
Affiliation(s)
- Marco Gallo
- Oncological Endocrinology Unit, Department of Medical Sciences, AOU Città della Salute e della Scienza di Torino, Via Genova 3, 10126, Turin, Italy.
| | - Luigi Gentile
- Endocrinology and Diabetes Unit, Cardinal Massaja Hospital, Asti, Italy
| | | | - Oscar Bertetto
- Head of the Piemonte and Valle d'Aosta Oncology Network, Turin, Italy
| | - Gennaro Clemente
- Institute of Food Science, National Research Council, IRPPS - Institute for Research on Population and Social Policies, Penta di Fisciano, Salerno, Italy
| |
Collapse
|
34
|
Samis J, Lee P, Zimmerman D, Arceci RJ, Suttorp M, Hijiya N. Recognizing Endocrinopathies Associated With Tyrosine Kinase Inhibitor Therapy in Children With Chronic Myelogenous Leukemia. Pediatr Blood Cancer 2016; 63:1332-8. [PMID: 27100618 DOI: 10.1002/pbc.26028] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 03/18/2016] [Indexed: 01/19/2023]
Abstract
Side effects of tyrosine kinase inhibitor (TKI) treatment vary in children and adults with chronic myelogenous leukemia (CML). As children have a much longer life expectancy than adults, TKI therapy may continue for decades and with long-term consequences that differ from adults. Children may develop endocrinopathies related to "off-target" effects of TKIs, such as delayed growth, changes in bone metabolism, thyroid abnormalities, and effects on puberty and fertility. These endocrinopathies present additional challenges for pediatric patients with CML. This review critically evaluates the literature on long-term endocrine side effects of TKIs in the pediatric CML population and provides suggested recommendations.
Collapse
Affiliation(s)
- Jill Samis
- Division of Pediatric Endocrinology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Paul Lee
- Division of Pediatric Hematology Oncology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| | - Donald Zimmerman
- Division of Pediatric Endocrinology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Robert J Arceci
- Department of Child Health, The Ron Matricaria Institute of Molecular Medicine at Phoenix Children's Hospital, University of Arizona College of Medicine, Phoenix, Arizona
| | - Meinolf Suttorp
- Pediatric Hematology, Oncology & Stem Cell Transplantation, Department of Pediatrics, Children's Hospital, Technical University of Dresden, Dresden, Germany
| | - Nobuko Hijiya
- Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, Illinois.,Division of Pediatric Hematology Oncology, Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois
| |
Collapse
|
35
|
Bernard V, Bouilly J, Kramer P, Carré N, Schlumberger M, Visser JA, Young J, Binart N. The Tyrosine Kinase Inhibitor Sunitinib Affects Ovulation but Not Ovarian Reserve in Mouse: A Preclinical Study. PLoS One 2016; 11:e0152872. [PMID: 27035144 PMCID: PMC4818017 DOI: 10.1371/journal.pone.0152872] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/21/2016] [Indexed: 12/30/2022] Open
Abstract
The aim of the study was to evaluate ovarian toxicity of tyrosine kinase inhibitor (TKI) sunitinib, since only scarce data are available on gonadal function after this treatment. Six-week-old female mice received orally, once daily, vehicle or sunitinib (50 mg/kg/d) during 5 weeks. Fertility parameters were analyzed from ovulation to litter assessment. Sunitinib exposure significantly reduced (i) corpora lutea number per ovary (1.1 ± 0.38 in sunitinib group versus 4 ± 0.79 in control group, p<0.01) and (ii) serum Anti Müllerian hormone (AMH) levels in sunitinib treated mice (12.01 ± 1.16) compared to control mice (14.33 ± 0.87 ng/ml, p< 0.05). However, primordial and growing follicles numbers per ovary were not different in both groups. After treatment withdrawal, female mice in both groups were able to obtain litters. These data could be helpful to counsel clinicians and patients, when fertility preservation methods are discussed, before TKI treatment in girls and young women.
Collapse
Affiliation(s)
- Valérie Bernard
- Inserm U1185, Univ. Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, 94276, France
| | - Justine Bouilly
- Inserm U1185, Univ. Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, 94276, France
| | - Piet Kramer
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Nadège Carré
- Inserm U1185, Univ. Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, 94276, France
| | - Martin Schlumberger
- Institut Gustave Roussy, Département de Médecine nucléaire et Oncologie endocrinienne, Université Paris-Sud, Villejuif, 94800, France
| | - Jenny A. Visser
- Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Jacques Young
- Inserm U1185, Univ. Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, 94276, France
- Service d’Endocrinologie et des Maladies de la Reproduction, Assistance Publique des Hôpitaux de Paris, Univ. Paris-Sud, Faculté de Médecine Paris-Sud, Le Kremlin Bicêtre, 94276, France
| | - Nadine Binart
- Inserm U1185, Univ. Paris-Sud, Université Paris Saclay, Le Kremlin-Bicêtre, 94276, France
- * E-mail:
| |
Collapse
|
36
|
Fountas A, Diamantopoulos LN, Tsatsoulis A. Tyrosine Kinase Inhibitors and Diabetes: A Novel Treatment Paradigm? Trends Endocrinol Metab 2015; 26:643-656. [PMID: 26492832 DOI: 10.1016/j.tem.2015.09.003] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 09/08/2015] [Accepted: 09/12/2015] [Indexed: 01/08/2023]
Abstract
Deregulation of protein tyrosine kinase (PTK) activity is implicated in various proliferative conditions. Multi-target tyrosine kinase inhibitors (TKIs) are increasingly used for the treatment of different malignancies. Recently, several clinical cases of the reversal of both type 1 and 2 diabetes mellitus (T1DM, T2DM) during TKI administration have been reported. Experimental in vivo and in vitro studies have elucidated some of the mechanisms behind this effect. For example, inhibition of Abelson tyrosine kinase (c-Abl) results in β cell survival and enhanced insulin secretion, while platelet-derived growth factor receptor (PDGFR) and epidermal growth factor receptor (EGFR) inhibition leads to improvement in insulin sensitivity. In addition, inhibition of vascular endothelial growth factor receptor 2 (VEGFR2) reduces the degree of islet cell inflammation (insulitis). Therefore, targeting several PTKs may provide a novel approach for correcting the pathophysiologic disturbances of diabetes.
Collapse
Affiliation(s)
- Athanasios Fountas
- Department of Endocrinology, University of Ioannina, Stavros Niarchos Avenue, 45110, Ioannina, Greece
| | | | - Agathocles Tsatsoulis
- Department of Endocrinology, University of Ioannina, Stavros Niarchos Avenue, 45110, Ioannina, Greece.
| |
Collapse
|
37
|
Anand D, Escalante CP. Ongoing Screening and Treatment to Potentially Reduce Tyrosine Kinase Inhibitor-Related Fatigue in Renal Cell Carcinoma. J Pain Symptom Manage 2015; 50:108-17. [PMID: 25701692 DOI: 10.1016/j.jpainsymman.2015.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 01/20/2015] [Accepted: 02/02/2015] [Indexed: 01/03/2023]
Abstract
CONTEXT Renal cell carcinoma (RCC) represents 1% to 4% of adult malignancies, and approximately 33% of patients with RCC present with metastatic disease and have a poor prognosis. Better understanding of RCC tumor biology has led to the development of several molecularly targeted agents, such as tyrosine kinase inhibitors (TKIs), to manage advanced disease. Although evolving data suggest these drugs may be beneficial in RCC, they are associated with significant toxicities. Cancer-related fatigue (CRF) is one of the most common toxicities associated with the TKIs used in RCC. OBJECTIVES To review the incidence, pathophysiology, and management of CRF in patients with RCC who are undergoing targeted therapy with TKIs. METHODS A comprehensive database search was performed using PubMed, Ovid, Embase, and MEDLINE. References of all cited articles also were reviewed. Data from articles published between 1975 and June 2014 were considered. A narrative review regarding the incidence, pathophysiology, and management of CRF in patients with RCC undergoing targeted therapy with TKIs was performed. RESULTS CRF is one of the most common TKI toxicities in patients with metastatic RCC and often is the dose-limiting toxicity. Management of TKI-related CRF can be difficult and may necessitate various nonpharmacologic and pharmacologic interventions. CONCLUSION TKI-related CRF in patients with RCC is a highly distressing complication of cancer therapy. CRF can substantially influence drug compliance, the ability to maximally treat, and quality of life. It is important to recognize this common, yet frequently underdiagnosed complication and initiate appropriate management strategies, to increase the likelihood for optimal outcomes.
Collapse
Affiliation(s)
- Deepa Anand
- Department of General Internal Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Carmen P Escalante
- Department of General Internal Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA.
| |
Collapse
|
38
|
Adrenocortical carcinoma: the management of metastatic disease. Crit Rev Oncol Hematol 2014; 92:123-32. [PMID: 24958272 DOI: 10.1016/j.critrevonc.2014.05.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 04/30/2014] [Accepted: 05/16/2014] [Indexed: 12/13/2022] Open
Abstract
Adrenocortical cancer is a rare malignancy. While surgery is the cornerstone of the management of localized disease, metastatic disease is hard to treat. Cytotoxic chemotherapy and mitotane have been utilized with a variable degree of benefit and few long-term responses. A growing understanding of the molecular pathogenesis of this malignancy as well as multidisciplinary and multi-institutional collaborative efforts will result in better defined targets and subsequently, effective novel therapies.
Collapse
|
39
|
Clinical pharmacokinetics of tyrosine kinase inhibitors: implications for therapeutic drug monitoring. Ther Drug Monit 2014; 35:562-87. [PMID: 24052062 DOI: 10.1097/ftd.0b013e318292b931] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The treatment of many malignancies has been improved in recent years by the introduction of molecular targeted therapies. These drugs interact preferentially with specific targets that are mutated and/or overexpressed in malignant cells. A group of such targets are the tyrosine kinases, against which a number of inhibitors (tyrosine kinase inhibitors, TKIs) have been developed. Imatinib, a TKI with targets that include the breakpoint cluster region-Abelson (bcr-abl) fusion protein kinase and mast/stem cell growth factor receptor kinase (c-Kit), was the first clinically successful drug of this type and revolutionized the treatment and prognosis of chronic myeloid leukemia and gastrointestinal stromal tumors. This success paved the way for the development of other TKIs for the treatment of a range of hematological malignancies and solid tumors. To date, 14 TKIs have been approved for clinical use and many more are under investigation. All these agents are given orally and are substrates of a range of drug transporters and metabolizing enzymes. In addition, some TKIs are capable of inhibiting their own transporters and metabolizing enzymes, making their disposition and metabolism at steady-state unpredictable. A given dose can therefore give rise to markedly different plasma concentrations in different patients, favoring the selection of resistant clones in the case of subtherapeutic exposure, and increasing the risk of toxicity if dosage is excessive. The aim of this review was to summarize current knowledge of the clinical pharmacokinetics and known adverse effects of the TKIs that are available for clinical use and to provide practical guidance on the implications of these data in patient management, in particular with respect to therapeutic drug monitoring.
Collapse
|
40
|
Santoni M, Conti A, Massari F, Arnaldi G, Iacovelli R, Rizzo M, De Giorgi U, Trementino L, Procopio G, Tortora G, Cascinu S. Treatment-related fatigue with sorafenib, sunitinib and pazopanib in patients with advanced solid tumors: an up-to-date review and meta-analysis of clinical trials. Int J Cancer 2014; 136:1-10. [PMID: 24415642 DOI: 10.1002/ijc.28715] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 12/23/2013] [Indexed: 12/24/2022]
Abstract
Fatigue is the most common symptom associated with cancer and cancer treatment. We performed an up-to-date meta-analysis to determine the incidence and relative risk (RR) of fatigue in patients (pts) with cancer treated with sorafenib (SO), sunitinib (SU) and pazopanib (PZ). PubMed databases were searched for articles published till August 2013. Eligible studies were selected according to PRISMA statement. Summary incidence, RR and 95% confidence intervals were calculated using random-effects or fixed-effects models based on the heterogeneity of selected studies. Fifteen studies were included in our analysis. A total of 6,996 pts was enrolled: 2,260 had renal cell carcinomas (RCC), 1,691 non-small cell lung cancers, 1,290 breast cancers, 823 hepatocellular carcinomas, 362 soft tissue sarcomas, 304 gastrointestinal solid tumors, 165 neuroendocrine tumors and 101 melanomas. When stratified by drug, SO registered lower incidence and RR of all and high-grade fatigue when compared to SU, whereas the difference between SO and PZ was significant only for all-grade fatigue (p < 0.001). The difference between SU and PZ was significant for high-grade (p < 0.001) but not for all-grade fatigue (p = 0.52). In RCC pts, PZ showed the lower incidence and RR of all and high-grade fatigue. The differences were significant for SU vs. SO (p < 0.001), SU vs. PZ (p < 0.001) and SO vs. PZ (p < 0.001). Treatment with SO, SU and PZ is associated with an increased incidence of fatigue in pts with cancer. Early and appropriate management is required to avoid unnecessary dose reductions and transitory or definitive treatment discontinuations.
Collapse
Affiliation(s)
- Matteo Santoni
- Medical Oncology, AOU Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Vergès B, Walter T, Cariou B. Endocrine side effects of anti-cancer drugs: effects of anti-cancer targeted therapies on lipid and glucose metabolism. Eur J Endocrinol 2014; 170:R43-55. [PMID: 24154684 DOI: 10.1530/eje-13-0586] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
During the past years, targeted therapies for cancer have been developed using drugs that have significant metabolic consequences. Among them, the mammalian target of rapamycin (mTOR) inhibitors and, to a much lesser extent, the tyrosine kinase inhibitors (TKIs) are involved. mTOR plays a key role in the regulation of cell growth as well as lipid and glucose metabolism. Treatment with mTOR inhibitors is associated with a significant increase in plasma triglycerides and LDL cholesterol. mTOR inhibitors seem to increase plasma triglycerides by reducing the activity of the lipoprotein lipase which is in charge of the catabolism of triglyceride-rich lipoproteins. The increase in LDL cholesterol observed with mTOR inhibitors seems to be due to a decrease in LDL catabolism secondary to a reduction of LDL receptor expression. In addition, treatment with mTOR inhibitors is associated with a high incidence of hyperglycemia, ranging from 13 to 50% in the clinical trials. The mechanisms responsible for hyperglycemia with new onset diabetes are not clear, but are likely due to the combination of impaired insulin secretion and insulin resistance. TKIs do not induce hyperlipidemia but alter glucose homeostasis. Treatment with TKIs may be associated either with hyperglycemia or hypoglycemia. The molecular mechanism by which TKIs control glucose homeostasis remains unknown. Owing to the metabolic consequences of these agents used as targeted anti-cancer therapies, a specific and personalized follow-up of blood glucose and lipids is recommended when using mTOR inhibitors and of blood glucose when using TKIs.
Collapse
Affiliation(s)
- Bruno Vergès
- Service Endocrinologie, Diabétologie et Maladies Métaboliques, INSERM CRI 866, Hôpital du Bocage, CHU Dijon, Université de Bourgogne, 21000 Dijon, France
| | | | | |
Collapse
|
42
|
Barnes N, Chemaitilly W. Endocrinopathies in survivors of childhood neoplasia. Front Pediatr 2014; 2:101. [PMID: 25295241 PMCID: PMC4172013 DOI: 10.3389/fped.2014.00101] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 09/06/2014] [Indexed: 01/26/2023] Open
Abstract
Advancements in cancer treatments have increased the number of survivors of childhood cancers. Endocrinopathies are common complications following cancer therapy and may occur decades later. The objective of the current review is to address the main endocrine abnormalities detected in childhood cancer survivors including disorders of the hypothalamic-pituitary axis, thyroid, puberty, gonads, bone, body composition, and glucose metabolism.
Collapse
Affiliation(s)
- Nicole Barnes
- Division of Pediatric Endocrinology, Department of Pediatric Medicine, St. Jude Children's Research Hospital , Memphis, TN , USA
| | - Wassim Chemaitilly
- Division of Pediatric Endocrinology, Department of Pediatric Medicine, St. Jude Children's Research Hospital , Memphis, TN , USA ; Epidemiology and Cancer Control, St. Jude Children's Research Hospital , Memphis, TN , USA
| |
Collapse
|
43
|
Abstract
Thyroid hormones (THs) may play a role in diseases other than hyper- and hypothyroidism. Several lines of evidence suggest tumor-promoting effects of TH and TH receptors. They are possibly mediated by phosphatidylinositol-3-kinase and MAPK and involve among others stimulation of angiogenesis via αvβ3. Thus, an increased risk for colon, lung, prostate, and breast cancer with lower TSH has been demonstrated in epidemiological studies, even suggesting a TH dose effect on cancer occurrence. Furthermore, higher TH levels were associated with an advanced clinical stage of breast and prostate cancer. In rodent models, TH stimulated growth and metastasis of tumor transplants, whereas hypothyroidism had opposite effects. In clinical studies of glioblastoma and head and neck cancer, hypothyroid patients showed longer survival than euthyroid patients. Also, patients with renal cell cancer that were treated with the tyrosine kinase inhibitor sunitinib and developed hypothyroidism in due course showed significantly longer survival than patients that remained euthyroid. Development of hypothyroidism was an independent predictor for survival in two studies. Yet, it is still possible that hypothyroidism is only a surrogate marker for treatment efficacy and does not positively influence treatment outcome by itself. Future cancer treatment studies, especially with substances that can induce hypothyroidism, should therefore be designed in a way that allows for an analysis of thyroid function status and its contribution on treatment outcome.
Collapse
Affiliation(s)
- Lars C Moeller
- Division of Laboratory Research, Department of Endocrinology and Metabolic Diseases, University of Duisburg-Essen, Hufelandstraße 55, 45127 Essen, Germany.
| | | |
Collapse
|
44
|
Abstract
CONTEXT The use of kinase inhibitors (KIs) in the treatment of cancer has become increasingly common, and practitioners must be familiar with endocrine-related side effects associated with these agents. This review provides an update to the clinician regarding the management of potential endocrinological effects of KIs. EVIDENCE ACQUISITION PubMed was employed to identify relevant manuscripts. A review of the literature was conducted, and data were summarized and incorporated. EVIDENCE SYNTHESIS KIs, including small molecule KIs and monoclonal antibodies directed against kinases, have emerged over the past decade as an important class of anticancer agents. KIs specifically interfere with signaling pathways that are dysregulated in certain types of cancers and also target common mechanisms of growth, invasion, metastasis, and angiogenesis. Currently, at least 20 KIs are approved as cancer therapeutics. However, KIs may affect a broad spectrum of targets and may have additional, unidentified mechanisms of action at the cellular level due to overlap between signaling pathways in the tumor cell and endocrine system. Recent reports in the literature have identified side effects associated with KIs, including alterations in thyroid function, bone metabolism, linear growth, gonadal function, fetal development, adrenal function, and glucose metabolism. CONCLUSIONS Clinicians need to monitor the thyroid functions of patients on KIs. In addition, bone density and vitamin D status should be assessed. Special care should be taken to follow linear growth and development in children taking these agents. Clinicians should counsel patients appropriately on the potential adverse effects of KIs on fetal development.
Collapse
Affiliation(s)
- Maya B Lodish
- Section on Endocrinology and Genetics, Program on Developmental Endocrinology and Genetics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, and the Pediatric Endocrinology Inter-Institute Training Program, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
45
|
Kawas LH, McCoy AT, Yamamoto BJ, Wright JW, Harding JW. Development of angiotensin IV analogs as hepatocyte growth factor/Met modifiers. J Pharmacol Exp Ther 2012; 340:539-48. [PMID: 22129598 DOI: 10.1124/jpet.111.188136] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
The 6-AH family [D-Nle-X-Ile-NH-(CH(2))(5)-CONH(2); where X = various amino acids] of angiotensin IV (Ang IV) analogs binds directly to hepatocyte growth factor (HGF) and inhibit HGF's ability to form functional dimers. The metabolically stabilized 6-AH family member, D-Nle-Tyr-Ile-NH-(CH(2))(5)-CONH(2,) had a t(1/2) in blood of 80 min compared with the parent compound norleual [Nle-Tyr-Leu-Ψ-(CH(2)-NH(2))(3-4)-His-Pro-Phe], which had a t(1/2) in blood of <5 min. 6-AH family members were found to act as mimics of the dimerization domain of HGF (hinge region) and inhibited the interaction of an HGF molecule with a (3)H-hinge region peptide resulting in an attenuated capacity of HGF to activate its receptor Met. This interference translated into inhibition of HGF-dependent signaling, proliferation, and scattering in multiple cell types at concentrations down into the low picomolar range. We also noted a significant correlation between the ability of the 6-AH family members to block HGF dimerization and inhibition of the cellular activity. Furthermore, a member of the 6-AH family with cysteine at position 2, was a particularly effective antagonist of HGF-dependent cellular activities. This compound suppressed pulmonary colonization by B16-F10 murine melanoma cells, which are characterized by an overactive HGF/Met system. Together, these data indicate that the 6-AH family of Ang IV analogs exerts its biological activity by modifying the activity of the HGF/Met system and offers the potential as therapeutic agents in disorders that are dependent on or possess an overactivation of the HGF/Met system.
Collapse
Affiliation(s)
- Leen H Kawas
- Department of Veterinary and Comparative, Anatomy, Pharmacology, and Physiology, PO Box 6520, Washington State University, Pullman, WA 99164-6520, USA
| | | | | | | | | |
Collapse
|
46
|
Kawas LH, Yamamoto BJ, Wright JW, Harding JW. Mimics of the dimerization domain of hepatocyte growth factor exhibit anti-Met and anticancer activity. J Pharmacol Exp Ther 2011; 339:509-18. [PMID: 21859930 DOI: 10.1124/jpet.111.185694] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The angiotensin IV analog norleual [Nle-Tyr-Leu-ψ-(CH(2)-NH(2))-Leu-His-Pro-Phe] has been shown recently to act as a hepatocyte growth factor (HGF)/Met antagonist capable of blocking the binding of HGF to the Met receptor, inhibiting HGF-dependent activation of Met, and attenuating HGF-dependent cellular activities. In addition, norleual exhibited marked anticancer activity. Homology between norleual and the dimerization domain (hinge region) of HGF led to the hypothesis that norleual acts by interfering with HGF dimerization/multimerization and functions as a dominant-negative hinge region mimic. To test this hypothesis we investigated the ability of norleual to bind to and inhibit the dimerization of HGF. To further evaluate the idea that norleual was acting as a hinge region mimic, we synthesized a hexapeptide representing the HGF hinge sequence and established its capacity to similarly block HGF-dependent activation of Met and HGF-dependent cellular functions. The hinge peptide not only bound with high affinity directly to HGF and blocked its dimerization but it also inhibited HGF-dependent Met activation, suppressed HGF-dependent cellular functions, and exhibited anticancer activity. The major implication of this study is that molecules targeting the dimerization domain of HGF may represent novel and viable anticancer therapeutic agents; the development of such molecules should be feasible using norleual and the hinge peptide as synthetic templates.
Collapse
Affiliation(s)
- Leen H Kawas
- Department of Veterinary and Comparative, Anatomy, Pharmacology, and Physiology, Washington State University, Pullman, WA 99164-6520, USA
| | | | | | | |
Collapse
|
47
|
Bauer AJ, Francis GL. Update on the molecular signature of differentiated thyroid cancer: clinical implications and potential opportunities. Expert Rev Endocrinol Metab 2011; 6:819-834. [PMID: 30780870 DOI: 10.1586/eem.11.68] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
With the development and maturation of new technologies, there has been a steady incorporation of powerful new tools into the evaluation and management of thyroid nodules and thyroid cancer. An increasing number of reports on oncogene testing and molecular screening in fine-needle aspiration biopsy samples have been published. However, there remains a paucity of data and consensus on combining both conventional and molecular technologies to determine the diagnosis and/or prognosis of disease. All patients with differentiated thyroid cancer stand to benefit from the identification and incorporation of reliable molecular markers into clinical practice. Identification of reliable markers would allow for stratification of treatment, affording the medical and surgical teams an ability to individually tailor evaluation and treatment, applying aggressive therapy and monitoring only when clinically warranted. For the majority of patients with thyroid cancer, the incorporation of a validated, multifaceted molecular profiling system may not improve survival; however, there is great opportunity for these efforts to decrease the morbidity associated with our current approach.
Collapse
Affiliation(s)
- Andrew J Bauer
- a Pediatric Endocrinology, Department of Pediatrics, Walter Reed Army Medical Center, Washington, DC, USA.
- b Uniformed Services University, Bethesda, MD, USA
- c Thyroid Center, Division of Endocrinology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Gary L Francis
- d Division of Endocrinology, Department of Pediatrics, Children's Hospital of Richmond at The Commonwealth University Health System, Medical College of Virginia, Richmond, VA, USA
| |
Collapse
|
48
|
Tanriverdi O, Unubol M, Taskin F, Meydan N, Sargin G, Guney E, Barutca S. Imatinib-associated bilateral gynecomastia and unilateral testicular hydrocele in male patient with metastatic gastrointestinal stromal tumor: a literature review. J Oncol Pharm Pract 2011; 18:303-10. [PMID: 22009701 DOI: 10.1177/1078155211424629] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Imatinib mesylate is a drug that has been approved for treatment of advanced gastrointestinal stromal tumors (GISTs) and patients with leukemia such as chronic myeloid or Philadelphia chromosome-positive acute lymphoblastic. Although it has been described only in one patient with testicular hydrocele and gynecomastia in the literature, several cases of male gynecomastia have been reported with the use of imatinib mesylate in chronic myeloid leukemia (GML). Generally, male mastoplasia resolves after discontinuation of imatinib treatment. We report a 73-year-old male with metastatic GISTs who developed gynecomastia and unilateral testicular hydrocele while receiving imatinib mesylate. Nine months after commencing imatinib treatment, gynecomastia and testicular hydrocele were determined. Hormone analyses requested showed serum testosterone levels below and serum estrogen levels above normal limits. During the first month after discontinuing imatinib mesylate treatment, serum testosterone level was normal and there was a partial regression in gynecomastia and testicular hydrocele. To our knowledge, this is the second report of male gynecomastia following imatinib mesylate treatment of a patient with GIST. In conclusion, male patients who are to receive treatment with imatinib mesylate may be monitored for serum testosterone levels and for other reproductive hormone profiles before initiation of the treatment and their breasts may be examined during follow-up visits.
Collapse
Affiliation(s)
- Ozgur Tanriverdi
- Department of Medical Oncology, Faculty of Medicine, Adnan Menderes University, Aydin, Turkey.
| | | | | | | | | | | | | |
Collapse
|
49
|
Hamnvik OPR, Larsen PR, Marqusee E. Thyroid dysfunction from antineoplastic agents. J Natl Cancer Inst 2011; 103:1572-87. [PMID: 22010182 DOI: 10.1093/jnci/djr373] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Unlike cytotoxic agents that indiscriminately affect rapidly dividing cells, newer antineoplastic agents such as targeted therapies and immunotherapies are associated with thyroid dysfunction. These include tyrosine kinase inhibitors, bexarotene, radioiodine-based cancer therapies, denileukin diftitox, alemtuzumab, interferon-α, interleukin-2, ipilimumab, tremelimumab, thalidomide, and lenalidomide. Primary hypothyroidism is the most common side effect, although thyrotoxicosis and effects on thyroid-stimulating hormone secretion and thyroid hormone metabolism have also been described. Most agents cause thyroid dysfunction in 20%-50% of patients, although some have even higher rates. Despite this, physicians may overlook drug-induced thyroid dysfunction because of the complexity of the clinical picture in the cancer patient. Symptoms of hypothyroidism, such as fatigue, weakness, depression, memory loss, cold intolerance, and cardiovascular effects, may be incorrectly attributed to the primary disease or to the antineoplastic agent. Underdiagnosis of thyroid dysfunction can have important consequences for cancer patient management. At a minimum, the symptoms will adversely affect the patient's quality of life. Alternatively, such symptoms can lead to dose reductions of potentially life-saving therapies. Hypothyroidism can also alter the kinetics and clearance of medications, which may lead to undesirable side effects. Thyrotoxicosis can be mistaken for sepsis or a nonendocrinologic drug side effect. In some patients, thyroid disease may indicate a higher likelihood of tumor response to the agent. Both hypothyroidism and thyrotoxicosis are easily diagnosed with inexpensive and specific tests. In many patients, particularly those with hypothyroidism, the treatment is straightforward. We therefore recommend routine testing for thyroid abnormalities in patients receiving these antineoplastic agents.
Collapse
Affiliation(s)
- Ole-Petter Riksfjord Hamnvik
- Division of Endocrinology, Diabetes, and Hypertension, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | | | |
Collapse
|
50
|
Uso de nuevas moléculas en el tratamiento del cáncer avanzado de tiroides. ACTA ACUST UNITED AC 2011; 58:381-6. [DOI: 10.1016/j.endonu.2011.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 07/19/2011] [Indexed: 11/19/2022]
|