51
|
Cardoso AM, Morais CM, Pena F, Marante T, Cunha PP, Jurado AS, Pedroso de Lima MC. Differentiation of glioblastoma stem cells promoted by miR-128 or miR-302a overexpression enhances senescence-associated cytotoxicity of axitinib. Hum Mol Genet 2021; 30:160-171. [PMID: 33438013 DOI: 10.1093/hmg/ddab011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/18/2020] [Accepted: 12/22/2020] [Indexed: 12/17/2022] Open
Abstract
Despite the intense global efforts towards an effective treatment of glioblastoma (GB), current therapeutic options are unsatisfactory with a median survival time of 12-15 months after diagnosis, which has not improved significantly over more than a decade. The high tumoral heterogeneity confers resistance to therapies, which has hindered a successful clinical outcome, GB remaining among the deadliest cancers. A hallmark of GB is its high recurrence rate, which has been attributed to the presence of a small subpopulation of tumor cells called GB stem-like cells (GSC). In the present work, the efficacy of a multimodal strategy combining microRNA (miRNA) modulation with new generation multitargeted tyrosine kinase inhibitors (imatinib and axitinib) was investigated aiming at tackling this subpopulation of GB cells. MiR-128 and miR-302a were selected as attractive therapeutic candidates on the basis of previous findings reporting that reestablishment of their decreased expression levels in GSC resulted in cell differentiation, which could represent a possible strategy to sensitize GSC to chemotherapy. Our results show that overexpression of miR-128 or miR-302a induced GSC differentiation, which enhanced senescence mediated by axitinib treatment, thus further impairing GSC proliferation. We also provided evidence for the capacity of GSC to efficiently internalize functionalized stable nucleic acid lipid particles, previously developed and successfully applied in our laboratory to target GB. Taken together, our findings will be important in the future design of a GB-targeted multimodal miRNA-based gene therapy, combining overexpression of miR-128 or miR-302a with axitinib treatment, endowed with the ability to overcome drug resistance.
Collapse
Affiliation(s)
- Ana M Cardoso
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, IIIUC-Institute for Interdisciplinary Research, Coimbra, Portugal
| | - Catarina M Morais
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, IIIUC-Institute for Interdisciplinary Research, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal Portugal
| | - Frederico Pena
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, IIIUC-Institute for Interdisciplinary Research, Coimbra, Portugal
| | - Tânia Marante
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, IIIUC-Institute for Interdisciplinary Research, Coimbra, Portugal
| | - Pedro P Cunha
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, IIIUC-Institute for Interdisciplinary Research, Coimbra, Portugal
| | - Amália S Jurado
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, IIIUC-Institute for Interdisciplinary Research, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal Portugal
| | - Maria C Pedroso de Lima
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, IIIUC-Institute for Interdisciplinary Research, Coimbra, Portugal
| |
Collapse
|
52
|
Ren BY, Dai XL, Wang J, Wu C, Lu TB, Chen JM. Cocrystallization of axitinib with carboxylic acids: preparation, crystal structures and dissolution behavior. CrystEngComm 2021. [DOI: 10.1039/d1ce00620g] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Three cocrystals of axitinib were prepared, and they demonstrated a significantly improved apparent solubility and dissolution rate without compromising physical stability.
Collapse
Affiliation(s)
- Bo-Ying Ren
- Institute for New Energy Materials and Low Carbon Technologies, School of Materials Science and Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Xia-Lin Dai
- Tianjin Key Laboratory of Drug Targeting and Bioimaging, School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Jie Wang
- Institute for New Energy Materials and Low Carbon Technologies, School of Materials Science and Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Chao Wu
- Tianjin Key Laboratory of Drug Targeting and Bioimaging, School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Tong-Bu Lu
- Institute for New Energy Materials and Low Carbon Technologies, School of Materials Science and Engineering, Tianjin University of Technology, Tianjin 300384, China
| | - Jia-Mei Chen
- Tianjin Key Laboratory of Drug Targeting and Bioimaging, School of Chemistry and Chemical Engineering, Tianjin University of Technology, Tianjin 300384, China
| |
Collapse
|
53
|
De Leo S, Trevisan M, Fugazzola L. Recent advances in the management of anaplastic thyroid cancer. Thyroid Res 2020; 13:17. [PMID: 33292371 PMCID: PMC7684758 DOI: 10.1186/s13044-020-00091-w] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/15/2020] [Indexed: 12/12/2022] Open
Abstract
Anaplastic thyroid cancer (ATC) is undoubtedly the thyroid cancer histotype with the poorest prognosis. The conventional treatment includes surgery, radiotherapy, and conventional chemotherapy. Surgery should be as complete as possible, securing the airway and ensuring access for nutritional support; the current standard of care of radiotherapy is the intensity-modulated radiation therapy; chemotherapy includes the use of doxorubicin or taxanes (paclitaxel or docetaxel) generally with platin (cisplatin or carboplatin). However, frequently, these treatments are not sufficient and a systemic treatment with kinase inhibitors is necessary. These include multitarget tyrosine kinase inhibitors (Lenvatinib, Sorafenib, Sunitinib, Vandetanib, Axitinib, Pazopanib, Pyrazolo-pyrimidine compounds), single target tyrosine kinase inhibitors (Dabrafenib plus Trametinib and Vemurafenib against BRAF, Gefitinib against EGFR, PPARγ ligands (e.g. Efatutazone), Everolimus against mTOR, vascular disruptors (e.g. Fosbretabulin), and immunotherapy (e.g. Spartalizumab and Pembrolizumab, which are anti PD-1/PD-L1 molecules). Therapy should be tailored to the patients and to the tumor genetic profile. A BRAF mutation analysis is mandatory, but a wider evaluation of tumor mutational status (e.g. by next-generation sequencing) is desirable. When a BRAFV600E mutation is detected, treatment with Dabrafenib and Trametinib should be preferred: this combination has been approved by the Food and Drug Administration for the treatment of patients with locally advanced or metastatic ATC with BRAFV600E mutation and with no satisfactory locoregional treatment options. Alternatively, Lenvatinib, regardless of mutational status, reported good results and was approved in Japan for treating unresectable tumors. Other single target mutation agents with fair results are Everolimus when a mutation involving the PI3K/mTOR pathway is detected, Imatinib in case of PDGF-receptors overexpression, and Spartalizumab in case of PD-L1 positive tumors. Several trials are currently evaluating the possible beneficial role of a combinatorial therapy in ATC. Since in this tumor several genetic alterations are usually found, the aim is to inhibit or disrupt several pathways: these combination strategies use therapy targeting angiogenesis, survival, proliferation, and may act against both MAPK and PI3K pathways. Investigating new treatment options is eagerly awaited since, to date, even the molecules with the best radiological results have not been able to provide a durable disease control.
Collapse
Affiliation(s)
- Simone De Leo
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Piazzale Brescia, 20, 20149, Milan, Italy.
| | - Matteo Trevisan
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Laura Fugazzola
- Department of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Piazzale Brescia, 20, 20149, Milan, Italy.,Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
54
|
Colombo C, Giancola N, Fugazzola L. Personalized treatment for differentiated thyroid cancer: current data and new perspectives. Minerva Endocrinol (Torino) 2020; 46:62-89. [PMID: 33213119 DOI: 10.23736/s2724-6507.20.03342-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
More conservative and personalized treatment options have been developed in recent years to face the rising diagnosis of low-risk differentiated thyroid carcinoma (DTC). The present review describes the change towards a more risk-adapted management either in the treatment or in the follow-up of DTC. Particular attention is given to the innovations introduced by the latest guidelines for low-risk tumors, starting from the most appropriate extension of surgery up to the postoperative management. The emerging role of active surveillance for low-risk microcarcinoma is discussed, as well as the development of percutaneous strategies in the setting of malignant thyroid disease. The recent use of approved new systemic target therapies for advanced radioiodine refractory thyroid cancer is reported, together with the description of new compounds in trial. Finally, we provide some considerations to improve the risk evaluation in a presurgical setting, especially related to the rising role of genetics, to enable better risk-based cancer management and personalized treatment choices.
Collapse
Affiliation(s)
- Carla Colombo
- Division of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy - .,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy -
| | - Noemi Giancola
- Division of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Laura Fugazzola
- Division of Endocrine and Metabolic Diseases, IRCCS Istituto Auxologico Italiano, Milan, Italy.,Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
55
|
Synthesis, in vitro, and in vivo evaluation of novel N-phenylindazolyl diarylureas as potential anti-cancer agents. Sci Rep 2020; 10:17969. [PMID: 33087745 PMCID: PMC7578069 DOI: 10.1038/s41598-020-74572-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 08/27/2020] [Indexed: 12/11/2022] Open
Abstract
Novel N-phenylindazole based diarylureas have been designed, synthesized and evaluated as potential anticancer agents. In vitro cell viability studies of these derivatives illustrate good potency with IC50 values in the range of 0.4–50 μM in several cancer cell lines including murine metastatic breast cancer 4T1, murine glioblastoma GL261, human triple negative breast cancer MDA-MB-231, human pancreatic cancer MIAPaCa-2, and human colorectal cancer cell line WiDr. The ester group in the lead compound 8i was modified to incorporate amino-amides to increase solubility and stability while retaining biological activity. Further in vitro studies reveal that lead candidates inhibit tube length in HUVEC cells. In vivo systemic toxicity studies indicate that these candidate compounds are well tolerated in mice without any significant side effects. Anticancer efficacy studies in WiDr tumor xenograft and 4T1 tumor syngraft models demonstrate that the lead candidate 11 exhibits significant antitumor properties as a single agent in these tumor models.
Collapse
|
56
|
Hazarika S, Barman P. Visible‐Light Cercosporin Catalyzed Sulfenylation of Electron‐Rich Compounds with Thiols under Transition‐Metal‐Free Conditions. ChemistrySelect 2020. [DOI: 10.1002/slct.202002512] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Sukanya Hazarika
- Department of Chemistry National Institute of Technology Silchar Assam 788010 India
| | - Pranjit Barman
- Department of Chemistry National Institute of Technology Silchar Assam 788010 India
| |
Collapse
|
57
|
The paradoxical roles of miR-4295 in human cancer: Implications in pathogenesis and personalized medicine. Genes Dis 2020; 9:638-647. [PMID: 35782974 PMCID: PMC9243315 DOI: 10.1016/j.gendis.2020.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 08/01/2020] [Accepted: 09/28/2020] [Indexed: 12/20/2022] Open
|
58
|
Hernando J, Ros J, Arroyo A, Capdevila J. Clinical and Translational Challenges in Thyroid Cancer. Curr Med Chem 2020; 27:4806-4822. [PMID: 32056516 DOI: 10.2174/0929867327666200214125712] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 01/23/2020] [Accepted: 01/31/2020] [Indexed: 01/15/2023]
Abstract
Thyroid cancer is the most common endocrine malignancy and it accounts for 1% of all newly diagnosed tumors. Approximately 10% of patients with differentiated thyroid carcinomas (DTC) and 30% with medullary thyroid carcinoma (MTC) could not be cured with locoregional treatment and could develop metastatic disease. In addition, one of the most aggressive solid tumors can arise from the thyroid gland, the anaplastic thyroid carcinoma, with a median overall survival of less than 6 months. Currently, only four drugs are approved for the treatment of DTC and MTC and several unmet needs are focusing the scientific discussions, including the resistant setting, the off-target side effects that may reduce the efficacy and the molecular knowledge-based combinations. In this review, we aimed to discuss the current molecular landscape and treatment of thyroid cancers, and the ongoing clinical and translational research lines focusing on new drugs and drug combinations to improve the inhibition of driver mutations, such as BRAF and RET, and how systemic therapies that improved outcomes of other cancer types, like immunotherapy and peptide receptor radionuclide therapy, may play a role in the future management of advanced thyroid cancers.
Collapse
Affiliation(s)
- Jorge Hernando
- Medical Oncology Department, Gastrointestinal and Endocrine Tumor Unit, Vall d´Hebron University Hospital, Vall Hebron Institute of Oncology (VHIO), Pg Vall d´Hebron 119-129, 08035 Barcelona, Spain
| | - Javier Ros
- Medical Oncology Department, Gastrointestinal and Endocrine Tumor Unit, Vall d´Hebron University Hospital, Vall Hebron Institute of Oncology (VHIO), Pg Vall d´Hebron 119-129, 08035 Barcelona, Spain
| | - Alvaro Arroyo
- Medical Oncology Department, Gastrointestinal and Endocrine Tumor Unit, Vall d´Hebron University Hospital, Vall Hebron Institute of Oncology (VHIO), Pg Vall d´Hebron 119-129, 08035 Barcelona, Spain
| | - Jaume Capdevila
- Medical Oncology Department, Gastrointestinal and Endocrine Tumor Unit, Vall d´Hebron University Hospital, Vall Hebron Institute of Oncology (VHIO), Pg Vall d´Hebron 119-129, 08035 Barcelona, Spain
| |
Collapse
|
59
|
Tappenden P, Carroll C, Hamilton J, Kaltenthaler E, Wong R, Wadsley J, Moss L, Balasubramanian S. Cabozantinib and vandetanib for unresectable locally advanced or metastatic medullary thyroid cancer: a systematic review and economic model. Health Technol Assess 2020; 23:1-144. [PMID: 30821231 DOI: 10.3310/hta23080] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Medullary thyroid cancer (MTC) is a rare form of cancer that affects patients' health-related quality of life (HRQoL) and survival. Cabozantinib (Cometriq®; Ipsen, Paris, France) and vandetanib (Caprelsa®; Sanofi Genzyme, Cambridge, MA, USA) are currently the treatment modality of choice for treating unresectable progressive and symptomatic MTC. OBJECTIVES (1) To evaluate the clinical effectiveness and safety of cabozantinib and vandetanib. (2) To estimate the incremental cost-effectiveness of cabozantinib and vandetanib versus each other and best supportive care. (3) To identify key areas for primary research. (4) To estimate the overall cost of these treatments in England. DATA SOURCES Peer-reviewed publications (searched from inception to November 2016), European Public Assessment Reports and manufacturers' submissions. REVIEW METHODS A systematic review [including a network meta-analysis (NMA)] was conducted to evaluate the clinical effectiveness and safety of cabozantinib and vandetanib. The economic analysis included a review of existing analyses and the development of a de novo model. RESULTS The systematic review identified two placebo-controlled trials. The Efficacy of XL184 (Cabozantinib) in Advanced Medullary Thyroid Cancer (EXAM) trial evaluated the efficacy and safety of cabozantinib in patients with unresectable locally advanced, metastatic and progressive MTC. The ZETA trial evaluated the efficacy and safety of vandetanib in patients with unresectable locally advanced or metastatic MTC. Both drugs significantly improved progression-free survival (PFS) more than the placebo (p < 0.001). The NMA suggested that, within the symptomatic and progressive MTC population, the effects on PFS were similar (vandetanib vs. cabozantinib: hazard ratio 1.14, 95% credible interval 0.41 to 3.09). Neither trial demonstrated a significant overall survival benefit for cabozantinib or vandetanib versus placebo, although data from ZETA were subject to potential confounding. Both cabozantinib and vandetanib demonstrated significantly better objective response rates and calcitonin (CTN) and carcinoembryonic antigen (CEA) response rates than placebo. Both cabozantinib and vandetanib produced frequent adverse events, often leading to dose interruption or reduction. The assessment group model indicates that, within the EU-label population (symptomatic and progressive MTC), the incremental cost-effectiveness ratios (ICERs) for cabozantinib and vandetanib are > £138,000 per quality-adjusted life-year (QALY) gained. Within the restricted EU-label population (symptomatic and progressive MTC with CEA/CTN doubling times of ≤ 24 months), the ICER for vandetanib is expected to be > £66,000 per QALY gained. The maximum annual budget impact within the symptomatic and progressive population is estimated to be ≈£2.35M for cabozantinib and ≈£5.53M for vandetanib. The costs of vandetanib in the restricted EU-label population are expected to be lower. LIMITATIONS The intention-to-treat populations of the EXAM and ZETA trials are notably different. The analyses of ZETA subgroups may be subject to confounding as a result of differences in baseline characteristics and open-label vandetanib use. Attempts to statistically adjust for treatment switching were unsuccessful. No HRQoL evidence was identified for the MTC population. CONCLUSIONS The identified trials suggest that cabozantinib and vandetanib improve PFS more than the placebo; however, significant OS benefits were not demonstrated. The economic analyses indicate that within the EU-label population, the ICERs for cabozantinib and vandetanib are > £138,000 per QALY gained. Within the restricted EU-label population, the ICER for vandetanib is expected to be > £66,000 per QALY gained. FUTURE RESEARCH PRIORITIES (1) Primary research assessing the long-term effectiveness of cabozantinib and vandetanib within relevant subgroups. (2) Reanalyses of the ZETA trial to investigate the impact of adjusting for open-label vandetanib use using appropriate statistical methods. (3) Studies assessing the impact of MTC on HRQoL. STUDY REGISTRATION This study is registered as PROSPERO CRD42016050403. FUNDING The National Institute for Health Research Health Technology Assessment programme.
Collapse
Affiliation(s)
- Paul Tappenden
- Health Economics and Decision Science, School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Christopher Carroll
- Health Economics and Decision Science, School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Jean Hamilton
- Health Economics and Decision Science, School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Eva Kaltenthaler
- Health Economics and Decision Science, School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | - Ruth Wong
- Health Economics and Decision Science, School of Health and Related Research (ScHARR), University of Sheffield, Sheffield, UK
| | | | | | - Sabapathy Balasubramanian
- Department of Oncology and Metabolism, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| |
Collapse
|
60
|
Bible KC, Menefee ME, Lin CC(J, Millward MJ, Maples WJ, Goh BC, Karlin NJ, Kane MA, Adkins DR, Molina JR, Donehower RC, Lim WT, Flynn PJ, Richardson RL, Traynor AM, Rubin J, LoRusso PM, Smallridge RC, Burton JK, Suman VJ, Kumar A, Voss JS, Rumilla KM, Kipp BR, Chintakuntlawar AV, Harris P, Erlichman C. An International Phase 2 Study of Pazopanib in Progressive and Metastatic Thyroglobulin Antibody Negative Radioactive Iodine Refractory Differentiated Thyroid Cancer. Thyroid 2020; 30:1254-1262. [PMID: 32538690 PMCID: PMC7482116 DOI: 10.1089/thy.2019.0269] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Introduction: Multikinase inhibitors have clinical activity in radioactive iodine refractory (RAIR) differentiated thyroid cancers (DTCs) but are not curative; optimal management and salvage therapies remain unclear. This study assessed clinical effects of pazopanib therapy in RAIR-DTC patients with progressive disease, examining in parallel biomarker that might forecast/precede therapeutic response. Methods: Assessment of responses and toxicities and of any association between thyroglobulin (Tg) changes cycle 1 and RECIST (response evaluation criteria in solid tumors) response to pazopanib therapy were prospectively undertaken in Tg antibody negative RAIR-DTC patients. RECIST progressive metastatic disease <6 months preceding enrollment was required. With a sample size of 68 (assuming 23 attaining partial response [PR]), there would be 90% chance of detecting a difference of >30% when the proportion of patients attaining PR whose Tg values decrease by >50% is >50% cycle 1 (one-sided α = 0.10, two sample test of proportions). Mean corpuscular volume (MCV) change or mutational status or pretreatment were also explored as early correlates of eventual RECIST response. Results: From 2009 to 2011, 60 individuals were treated and evaluated; (one additional patient withdrew; another was found ineligible before therapy initiation); 91.7% had previous systemic therapy beyond RAI. Adverse events included one death (thromboembolic) deemed possibly pazopanib associated. Twenty-two confirmed RECIST PRs resulted (36.7%, confidence interval; CI [24.6-50.1]); mean administered 4-week cycles was 10. Among 44 fully accessible patients, the Tg nadir was greater among the 20 attaining PR (median: -86.8%; interquartile range [IQR]: -90.7% to -70.9%) compared with the 28 who did not (median: -69.0%; IQR: -78.1% to -27.7%, Wilcoxon rank-sum test: p = 0.002). However, the difference in the proportion of PRs among those whose Tg fell ≥50% after cycle 1 versus those that did not were not significantly correlated (-23.5% [CI: -55.3 to 8.3]; Fisher's exact test p-value = 0.27). RECIST response was also not correlated with/predicted by early MCV change, receipt of prior therapy, or tumor mutational status. Conclusions: This trial prospectively confirmed pazopanib to have clinical activity and manageable toxicities in patients with progressive RAIR-DTC. Response to pazopanib, however, was not robustly forecast by early associated changes in Tg or MCV, by prior therapy, or by tumor mutational status. ClinicalTrials.gov NCT00625846.
Collapse
Affiliation(s)
- Keith C. Bible
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
- Address correspondence to: Keith C. Bible, MD, PhD, Division of Medical Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA
| | - Michael E. Menefee
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | | | - Michael J. Millward
- Department of Medical Oncology, University of Western Australia, Perth, Australia
| | - William J. Maples
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Boon Cher Goh
- Division of Medical Oncology, National University Cancer Institute, Singapore, Singapore
| | - Nina J. Karlin
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Madeleine A. Kane
- Department of Medical Oncology, University of Colorado, Denver, Colorado, USA
| | - Douglas R. Adkins
- Division of Medical Oncology, Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Julian R. Molina
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ross C. Donehower
- Division of Medical Oncology, Department of Medicine, Johns Hopkins, Baltimore, Maryland, USA
| | - Wan-Teck Lim
- Division of Medical Oncology, National University Cancer Institute, Singapore, Singapore
| | | | - Ronald L. Richardson
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Anne M. Traynor
- Division of Hematology, Medical Oncology and Palliative Care, Department of Medicine, University of Wisconsin, Madison, Wisconsin, USA
| | - Joseph Rubin
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Robert C. Smallridge
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Jill K. Burton
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Vera J. Suman
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Aditi Kumar
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jessie S. Voss
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Kandalaria M. Rumilla
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Benjamin R. Kipp
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Pamela Harris
- Cancer Therapy Evaluation Program (CTEP), Bethesda, Maryland, USA
| | - Charles Erlichman
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
61
|
Abstract
The diagnostic modalities, stratification tools, and treatment options for patients with thyroid cancer have rapidly evolved since the development of the American Thyroid Association (ATA) guidelines in 2015. This review compiles newer concepts in diagnosis, stratification tools and treatment options for patients with differentiated thyroid cancer (DTC), medullary thyroid carcinoma (MTC) and anaplastic thyroid cancer (ATC). Newer developments apply precision medicine in thyroid cancer patients to avoid over-treatment in low risk disease and under-treatment in high risk disease. Among novel patient-tailored therapies are selective RET inhibitors that have shown efficacy in the treatment of MTC with limited systemic toxicity compared with non-specific tyrosine kinase inhibitors. The combination of BRAF and MEK inhibitors have revolutionized management of BRAF V600E mutant ATC. Several immunotherapeutic agents are being actively investigated in the treatment of all forms of thyroid cancer. In this review, we describe the recent advances in the diagnosis and management of DTC, MTC, and ATC, with an emphasis on novel treatment modalities.
Collapse
Affiliation(s)
- Katherine A. Araque
- Endocrinology Department, Pacific Neuroscience Institute, John Wayne Cancer Institute, Santa Monica, CA, USA
| | - Sriram Gubbi
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Joanna Klubo-Gwiezdzinska
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), National Institutes of Health (NIH), Bethesda, MD, USA
| |
Collapse
|
62
|
Al-Jundi M, Thakur S, Gubbi S, Klubo-Gwiezdzinska J. Novel Targeted Therapies for Metastatic Thyroid Cancer-A Comprehensive Review. Cancers (Basel) 2020; 12:E2104. [PMID: 32751138 PMCID: PMC7463725 DOI: 10.3390/cancers12082104] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 07/22/2020] [Accepted: 07/25/2020] [Indexed: 12/18/2022] Open
Abstract
The knowledge on thyroid cancer biology has grown over the past decade. Thus, diagnostic and therapeutic strategies to manage thyroid cancer are rapidly evolving. With new insights into tumor biology and cancer genetics, several novel therapies have been approved for the treatment of thyroid cancer. Tyrosine kinase inhibitors (TKIs), such as lenvatinib and sorafenib, have been successfully utilized for the treatment of radioactive iodine (RAI)-refractory metastatic differentiated thyroid cancer (DTC). In addition, pretreatment with mitogen-activated protein kinase (MAPK) inhibitors (trametinib and selumetinib) has been shown to restore RAI avidity in previously RAI-refractory DTCs. Local therapies, such as external beam radiation and radiofrequency/ethanol ablation, have also been employed for treatment of DTC. Vandetanib and cabozantinib are the two TKIs currently approved by the Food and Drug Administration (FDA) for the treatment of medullary thyroid cancer (MTC). Other novel therapies, such as peptide receptor radionuclide therapy and carcinoembryonic antigen (CEA) vaccine, have also been utilized in treating MTC. Ongoing trials on selective rearranged-during-transfection (RET) protooncogene inhibitors, such as LOXO-292 and BLU-667, have demonstrated promising results in the treatment of metastatic MTC resistant to non-selective TKIs. The FDA-approved BRAF/MEK inhibitor combination of dabrafenib and trametinib has revolutionized treatment of BRAFV600E mutation positive anaplastic thyroid cancer. Several other emerging classes of medications, such as gene fusion inhibitors and immune checkpoint inhibitors, are being actively investigated in several clinical trials. In this review, we describe the molecular landscape of thyroid cancer and novel targeted therapies and treatment combinations available for the treatment of metastatic thyroid cancer.
Collapse
Affiliation(s)
| | | | | | - Joanna Klubo-Gwiezdzinska
- Thyroid Tumors and Functional Thyroid Disorders Section, Metabolic Disease Branch, National Institute of Diabetes, Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20814, USA; (M.A.-J.); (S.T.); (S.G.)
| |
Collapse
|
63
|
Lin CY, Chang JS, Huang SM, Hung CJ, Hung CL, Chang CT, Yang HR, Hsieh TC, Huang YH, Tsai HJ. Experience of sorafenib treatment in differentiated thyroid cancer from Taiwan. J Formos Med Assoc 2020; 120:189-195. [PMID: 32402521 DOI: 10.1016/j.jfma.2020.04.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/17/2020] [Accepted: 04/15/2020] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Sorafenib has been shown to prolong the progression free survival (PFS) of advanced radioiodine (RAI) refractory differentiated thyroid cancer (DTC) and has been approved by the FDA as the result of the phase III DECISION trial. Sorafenib has been reimbursed for the treatment of RAI refractory DTC in Taiwan since Jan 2017. High percentage of adverse events (AE) was noted in DECISION trial. We conducted a study to show the real-world experience of sorafenib in Taiwan. METHODS We retrospectively collected the clinical data, including dose, AE, and PFS of sorafenib, of the DTC patients who received sorafenib treatment in National Cheng Kung University Hospital and China Medical University Hospital by chart review from 2012 to 2018. RESULTS Thirty-six advanced DTC patients with progression were included in this study. The starting dose of sorafenib in most patients was 200 mg twice daily and the mean daily maintenance dose was 433 mg. Five patients had partial response (13.9%) and 28 patients had stable disease (77.8%). The median PFS was 17.3 months (95% confidence interval: 11.9-33.6 months). Daily maintenance dose ≥ 600 mg was associated with better PFS (median PFS, not reached). The most common toxicity of sorafenib was hand foot skin reaction (69%), followed by diarrhea (42%), and skin rash (33%). Most of the toxicities were grade I/II. CONCLUSION Higher maintenance dose of sorafenib is associated with longer PFS while starting from half dose is feasible to minimize the incidence of high grade toxicities in the real-world use of sorafenib.
Collapse
Affiliation(s)
- Chen-Yuan Lin
- Division of Hematology and Oncology, China Medical University Hospital, Taichung, Taiwan; School of Pharmacy, China Medical University, Taichung, Taiwan
| | - Jeffrey S Chang
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan
| | - Shih-Ming Huang
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Asian International Thyroid Center, Chang Bing Show Chwan Memorial Hospital, Lugang Town, Changhua County, Taiwan
| | - Chung-Jye Hung
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Surgery, Tainan Hospital, Ministry of Health and Welfare, Tainan, Taiwan
| | - Chien-Ling Hung
- Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chwen-Tzuei Chang
- Division of Metabolism, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan; Department of Internal Medicine, College of Medicine, China Medical University, Taichung, Taiwan
| | - Horng-Ren Yang
- Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan; Department of Surgery, China Medical University Hospital, Taichung, Taiwan; Department of Surgery, College of Medicine, China Medical University, Taichung, Taiwan
| | - Te-Chun Hsieh
- Department of Nuclear Medicine, China Medical University Hospital, Taichung, Taiwan; Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung, Taiwan
| | - Yu-Hui Huang
- Nursing Department of National Cheng Kung University Hospital, Tainan, Taiwan
| | - Hui-Jen Tsai
- National Institute of Cancer Research, National Health Research Institutes, Tainan, Taiwan; Division of Hematology and Oncology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Division of Hematology and Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
| |
Collapse
|
64
|
Subbiah V, Cote GJ. Advances in Targeting RET-Dependent Cancers. Cancer Discov 2020; 10:498-505. [PMID: 32094155 PMCID: PMC7125013 DOI: 10.1158/2159-8290.cd-19-1116] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/04/2019] [Accepted: 01/28/2020] [Indexed: 12/29/2022]
Abstract
RET alterations have been characterized as oncogenic drivers in multiple cancers. The clinical validation of highly selective RET inhibitors demonstrates the utility of specific targeting of aberrantly activated RET in patients with cancers such as medullary thyroid cancer or non-small cell lung cancer. The remarkable responses observed have opened the field of RET-targeted inhibitors. In this review, we seek to focus on the impact of therapeutic RET targeting in cancers. SIGNIFICANCE: Successful clinical translation of selective RET inhibitors is poised to alter the therapeutic landscape of altered cancers. Questions that clearly need to be addressed relate to the ability to maintain long-term inhibition of tumor cell growth, how to prepare for the potential mechanisms of acquired resistance, and the development of next-generation selective RET inhibitors.
Collapse
Affiliation(s)
- Vivek Subbiah
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, Texas
- MD Anderson Cancer Network, Houston, Texas
| | - Gilbert J Cote
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
65
|
Li K, Zhan Y, Shen X. Multiple intracranial lesions with lung adenocarcinoma: A rare case of MOG-IgG-associated encephalomyelitis. Mult Scler Relat Disord 2020; 42:102064. [PMID: 32234600 DOI: 10.1016/j.msard.2020.102064] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 03/17/2020] [Accepted: 03/20/2020] [Indexed: 10/24/2022]
Abstract
BACKGROUND Myelin oligodendrocyte glycoprotein (MOG) is an important marker on the surface of oligodendrocytes and is associated with many demyelinating diseases. Recently, MOG-IgG-associated encephalomyelitis (MOG-EM) has been proposed as a disease entity with a preliminary diagnosis standard. Some patients with lung cancer have been reported to be seropositive for onconeural antibodies; however, lung cancer cases with MOG-EM have not been previously reported. METHODS We report the case of a patient with lung adenocarcinoma with multiple intracranial lesions found during molecular targeted therapy. RESULTS The patient tested positive for MOG antibody in her cerebrospinal fluid, and the therapeutic effect of steroids was excellent. CONCLUSION This is the first reported case of MOG-EM coincident with lung cancer in a patient with multiple intracranial lesions. When patients present with a history of malignant tumors or suspected paraneoplastic neurological syndrome, clinicians should also be alert to the presence of other autoimmune antibodies such as MOG-IgG to avoid treatment delay.
Collapse
Affiliation(s)
- Ke Li
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Yajing Zhan
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Xiangmin Shen
- Department of Neurology, The Second Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
66
|
Abstract
OPINION STATEMENT The landscape of treatment options for radioactive iodine refractory thyroid cancer is rapidly changing. While there are no curative options in this setting, tyrosine kinase inhibitors (TKIs) have revolutionized the management of radioiodine refractory disease to help delay progression of metastatic and life-threatening disease. Ongoing development of more selective targeted inhibitors will certainly improve medication tolerability and tumor specificity. In this review, we discuss the epidemiology of radioactive iodine refractory thyroid cancer and examine the definition of radioactive iodine refractory disease and the current systemic therapy options. We then discuss molecularly targeted strategies both approved by the FDA and currently under study in clinical trials. In particular, we examine the data relevant to specific targeted mutations in thyroid cancer. We also discuss novel approaches in development, such as immunotherapy, to the management of radioactive iodine refractory disease.
Collapse
Affiliation(s)
- Kedar Kirtane
- Department of Oncology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA.
| | - Mara Y Roth
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington and Seattle Cancer Care Alliance, Seattle, WA, USA
| |
Collapse
|
67
|
Exploring receptor tyrosine kinases-inhibitors in Cancer treatments. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2019. [DOI: 10.1186/s43042-019-0035-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
AbstractBackgroundReceptor tyrosine kinases (RTKs) are signaling enzymes responsible for the transfer of Adenosine triphosphate (ATP) γ-phosphate to the tyrosine residues substrates. RTKs demonstrate essential roles in cellular growth, metabolism, differentiation, and motility. Anomalous expression of RTK customarily leads to cell growth dysfunction, which is connected to tumor takeover, angiogenesis, and metastasis. Understanding the structure, mechanisms of adaptive and acquired resistance, optimizing inhibition of RTKs, and eradicating cum minimizing the havocs of quiescence cancer cells is paramount.MainTextTyrosine kinase inhibitors (TKIs) vie with RTKs ATP-binding site for ATP and hitherto reduce tyrosine kinase phosphorylation, thus hampering the growth of cancer cells. TKIs can either be monoclonal antibodies that compete for the receptor’s extracellular domain or small molecules that inhibit the tyrosine kinase domain and prevent conformational changes that activate RTKs. Progression of cancer is related to aberrant activation of RTKs due to due to mutation, excessive expression, or autocrine stimulation.ConclusionsUnderstanding the modes of inhibition and structures of RTKs is germane to the design of novel and potent TKIs. This review shed light on the structures of tyrosine kinases, receptor tyrosine kinases, tyrosine kinase inhibitors, minimizing imatinib associated toxicities, optimization of tyrosine kinase inhibition in curtailing quiescence in cancer cells and the prospects of receptor tyrosine kinase based treatments.
Collapse
|
68
|
Filetti S, Durante C, Hartl D, Leboulleux S, Locati LD, Newbold K, Papotti MG, Berruti A. Thyroid cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up†. Ann Oncol 2019; 30:1856-1883. [PMID: 31549998 DOI: 10.1093/annonc/mdz400] [Citation(s) in RCA: 677] [Impact Index Per Article: 112.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
MESH Headings
- Humans
- Adenocarcinoma, Follicular/diagnosis
- Adenocarcinoma, Follicular/epidemiology
- Adenocarcinoma, Follicular/pathology
- Adenocarcinoma, Follicular/therapy
- Carcinoma, Neuroendocrine/diagnosis
- Carcinoma, Neuroendocrine/epidemiology
- Carcinoma, Neuroendocrine/pathology
- Carcinoma, Neuroendocrine/therapy
- Follow-Up Studies
- Thyroid Cancer, Papillary/diagnosis
- Thyroid Cancer, Papillary/epidemiology
- Thyroid Cancer, Papillary/pathology
- Thyroid Cancer, Papillary/therapy
- Thyroid Carcinoma, Anaplastic/diagnosis
- Thyroid Carcinoma, Anaplastic/epidemiology
- Thyroid Carcinoma, Anaplastic/pathology
- Thyroid Carcinoma, Anaplastic/therapy
- Thyroid Neoplasms/diagnosis
- Thyroid Neoplasms/epidemiology
- Thyroid Neoplasms/pathology
- Thyroid Neoplasms/therapy
Collapse
Affiliation(s)
- S Filetti
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - C Durante
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - D Hartl
- Department of Head and Neck Oncology, Gustave Roussy, Villejuif; Université Paris Saclay, Villejuif
| | - S Leboulleux
- Université Paris Saclay, Villejuif; Department of Nuclear Medicine and Endocrine Oncology, Gustave Roussy, Villejuif, France
| | - L D Locati
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - K Newbold
- Head and Neck Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - M G Papotti
- Department of Pathology, University of Turin, Turin
| | - A Berruti
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Medical Oncology Unit, University of Brescia, ASST Spedali Civili, Brescia, Italy
| |
Collapse
|
69
|
Cabanillas ME, Ryder M, Jimenez C. Targeted Therapy for Advanced Thyroid Cancer: Kinase Inhibitors and Beyond. Endocr Rev 2019; 40:1573-1604. [PMID: 31322645 PMCID: PMC7341904 DOI: 10.1210/er.2019-00007] [Citation(s) in RCA: 225] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 07/11/2019] [Indexed: 02/07/2023]
Abstract
The treatment of advanced thyroid cancer has undergone rapid evolution in the last decade, with multiple kinase inhibitor drug approvals for each subtype of thyroid cancer and a number of other commercially available drugs that have been studied for this indication. Although most of the US Food and Drug Administration (FDA)-approved drugs are antiangiogenic multikinase inhibitors-vandetanib, cabozantinib, sorafenib, lenvatinib-there are two FDA indications that are mutation specific-dabrafenib/trametinib for BRAF-mutated anaplastic thyroid cancer and larotrectinib for NTRK-fusion thyroid cancer. Furthermore, other mutation-specific drugs, immunotherapies, and novel strategies for advanced thyroid cancer are under investigation. Understanding the molecular basis of thyroid cancer, the drugs of interest for treatment of advanced thyroid cancer, and how these drugs can be administered safely and in the appropriate clinical scenario are the topics of this review.
Collapse
Affiliation(s)
- Maria E Cabanillas
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mabel Ryder
- Department of Endocrinology and Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Camilo Jimenez
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
70
|
Aashiq M, Silverman DA, Na'ara S, Takahashi H, Amit M. Radioiodine-Refractory Thyroid Cancer: Molecular Basis of Redifferentiation Therapies, Management, and Novel Therapies. Cancers (Basel) 2019; 11:E1382. [PMID: 31533238 PMCID: PMC6770909 DOI: 10.3390/cancers11091382] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/11/2019] [Accepted: 09/12/2019] [Indexed: 12/13/2022] Open
Abstract
Recurrent, metastatic disease represents the most frequent cause of death for patients with thyroid cancer, and radioactive iodine (RAI) remains a mainstay of therapy for these patients. Unfortunately, many thyroid cancer patients have tumors that no longer trap iodine, and hence are refractory to RAI, heralding a poor prognosis. RAI-refractory (RAI-R) cancer cells result from the loss of thyroid differentiation features, such as iodide uptake and organification. This loss of differentiation features correlates with the degree of mitogen-activated protein kinase (MAPK) activation, which is higher in tumors with BRAF (B-Raf proto-oncogene) mutations than in those with RTK (receptor tyrosine kinase) or RAS (rat sarcoma) mutations. Hence, inhibition of the mitogen-activated protein kinase kinase-1 and -2 (MEK-1 and -2) downstream of RAF (rapidly accelerated fibrosarcoma) could sensitize RAI refractivity in thyroid cancer. However, a significant hurdle is the development of secondary tumor resistance (escape mechanisms) to these drugs through upregulation of tyrosine kinase receptors or another alternative signaling pathway. The sodium iodide symporter (NIS) is a plasma membrane glycoprotein, a member of solute carrier family 5A (SLC5A5), located on the basolateral surfaces of the thyroid follicular epithelial cells, which mediates active iodide transport into thyroid follicular cells. The mechanisms responsible for NIS loss of function in RAI-R thyroid cancer remains unclear. In a study of patients with recurrent thyroid cancer, expression levels of specific ribosomal machinery-namely PIGU (phosphatidylinositol glycan anchor biosynthesis class U), a subunit of the GPI (glycosylphosphatidylinositol transamidase complex-correlated with RAI avidity in radioiodine scanning, NIS levels, and biochemical response to RAI treatment. Here, we review the proposed mechanisms for RAI refractivity and the management of RAI-refractive metastatic, recurrent thyroid cancer. We also describe novel targeted systemic agents that are in use or under investigation for RAI-refractory disease, their mechanisms of action, and their adverse events.
Collapse
Affiliation(s)
- Mohamed Aashiq
- Department of Head and Neck Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Deborah A Silverman
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Shorook Na'ara
- Department of Otolaryngology, Head and Neck Surgery, and the Laboratory for Applied Cancer Research, Rappaport Institute of Medicine and Research, The Technion, Israel Institute of Technology, Haifa 3109601, Israel.
| | - Hideaki Takahashi
- Department of Head and Neck Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Moran Amit
- Department of Head and Neck Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
71
|
Khatami F, Larijani B, Nikfar S, Hasanzad M, Fendereski K, Tavangar SM. Personalized treatment options for thyroid cancer: current perspectives. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2019; 12:235-245. [PMID: 31571972 PMCID: PMC6750856 DOI: 10.2147/pgpm.s181520] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 06/14/2019] [Indexed: 12/16/2022]
Abstract
Thyroid cancer is one of the most common endocrine malignancies, with increasing incidence all over the world. In spite of good prognosis for differentiated thyroid carcinoma, for an unknown reason, about 5–10% of the patients, the cancer will show aggressive behavior, develop metastasis, and be refractory to treatment strategies like radioactive iodine. Regarding the genetic information, each thyroid cancer patient can be considered as an individual unique one, with unique genetic information. Contrary to standard chemotherapy drugs, target therapy components aim at one or more definite molecular pathway on cancer cells, so their selection is underlying patient’s genetic information. Nowadays, several mutations and rearrangements including BRAF, VEGF receptors, RET, and RET/PTC, KDR, KIT, PDGFRA, CD274, and JAK2 are taken into account for the therapeutic components like larotrectinib (TRK inhibitor), vemurafenib, sunitinib, sorafenib, selumetinib, and axitinib. With the new concept of personalized treatment of thyroid cancer diagnoses, planning treatment, finding out how well treatment will work, and estimating a prognosis has changed for the better over the last decade.
Collapse
Affiliation(s)
- Fatemeh Khatami
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shekoufeh Nikfar
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mandana Hasanzad
- Personalized Medicine Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Medical Genomics Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kiarad Fendereski
- Pediateric Urology and Regenerative Medicine Research Center, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Mohammad Tavangar
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pathology, Dr. Shariati Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
72
|
Molecular Alterations in Thyroid Cancer: From Bench to Clinical Practice. Genes (Basel) 2019; 10:genes10090709. [PMID: 31540307 PMCID: PMC6771012 DOI: 10.3390/genes10090709] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/26/2019] [Accepted: 09/10/2019] [Indexed: 12/11/2022] Open
Abstract
Thyroid cancer comprises different clinical and histological entities. Whereas differentiated (DTCs) malignancies are sensitive to radioiodine therapy, anaplastic (ATCs) and medullary (MTCs) tumors do not uptake radioactive iodine and display aggressive features associated with a poor prognosis. Moreover, in a majority of DTCs, disease evolution leads to the progressive loss of iodine sensitivity. Hence, iodine-refractory DTCs, along with ATCs and MTCs, require alternative treatments reflective of their different tumor biology. In the last decade, the molecular mechanisms promoting thyroid cancer development and progression have been extensively studied. This has led to a better understanding of the genomic landscape, displayed by thyroid malignancies, and to the identification of novel therapeutic targets. Indeed, several pharmacological compounds have been developed for iodine-refractory tumors, with four multi-target tyrosine kinase inhibitors already available for DTCs (sorafenib and lenvatinib) and MTCs (cabozantib and vandetanib), and a plethora of drugs currently being evaluated in clinical trials. In this review, we will describe the genomic alterations and biological processes intertwined with thyroid cancer development, also providing a thorough overview of targeted drugs already tested or under investigation for these tumors. Furthermore, given the existing preclinical evidence, we will briefly discuss the potential role of immunotherapy as an additional therapeutic strategy for the treatment of thyroid cancer.
Collapse
|
73
|
Sherman EJ, Dunn LA, Schöder H, Ho AL, Baxi SS, Ghossein RA, Haque SS, Sima C, Tuttle RM, Pfister DG. Phase 2 study of vascular endothelial growth factor trap for the treatment of metastatic thyroid cancer. Cancer 2019; 125:2984-2990. [PMID: 31174237 DOI: 10.1002/cncr.32046] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/04/2019] [Accepted: 02/08/2019] [Indexed: 12/27/2022]
Abstract
BACKGROUND Several multitargeted tyrosine kinase inhibitors (TKIs) have demonstrated activity in patients with thyroid cancer that is refractory to radioactive iodine (RAI). The antitumor effect is attributed at least in part to the ability of these TKIs to inhibit angiogenesis in these vascular tumors. Vascular endothelial growth factor (VEGF) Trap (VT) is a recombinantly produced fusion protein consisting solely of human sequences for VEGF receptors 1 and 2 extracellular domains and human immunoglobulin 1. Evaluating VT in patients with thyroid cancer is reasonable considering the activity observed with TKIs targeting VEGF. METHODS The current study was a single-institution, phase 2, Simon 2-stage design (21 to >41 patients) study based on the objective response rate and/or 6-month progression-free survival as the primary endpoints. Eligible patients were required to have progressive, RAI-refractory and/or [18 F]fludeoxyglucose-avid, recurrent and/or metastatic, nonmedullary, nonanaplastic thyroid cancer; disease that was measurable using Response Evaluation Criteria In Solid Tumors (RECIST) criteria; and adequate organ and bone marrow function. VT at a dose of 4 mg/kg intravenously was administered every 14 days. RESULTS A total of 40 patients were included in the analysis. Of these patients, 24 had papillary thyroid cancer, 2 had follicular thyroid cancer, and 11 had Hurthle cell thyroid cancer. The final 3 tumors were classified as poorly differentiated. There were no complete and/or partial responses noted; 34 patients achieved stable disease and 6 patients experienced disease progression as their best response. Of the 34 patients with stable disease, 16 remained on the study for >6 months and 6 patients remained on the study for >12 months. The median duration on treatment was 4.1 months (range, 0.6-30.8 months). CONCLUSIONS Unlike TKIs, which have shown responses in this setting, to the authors' knowledge there have been no responses observed with the use of single-agent VT to date. It does not appear to be a promising drug for the treatment of patients with thyroid cancer.
Collapse
Affiliation(s)
- Eric J Sherman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Lara A Dunn
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Heiko Schöder
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Alan L Ho
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Shrujal S Baxi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Ronald A Ghossein
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sofia S Haque
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Cami Sima
- Department of Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Robert Michael Tuttle
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Cornell Medicine, New York, New York
| | - David G Pfister
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Medicine, Weill Cornell Medicine, New York, New York
| |
Collapse
|
74
|
Jerkovich F, García Falcone MG, Pitoia F. The experience of an Endocrinology Division on the use of tyrosine multikinase inhibitor therapy in patients with radioiodine-resistant differentiated thyroid cancer. Endocrine 2019; 64:632-638. [PMID: 30820748 DOI: 10.1007/s12020-019-01883-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 02/22/2019] [Indexed: 12/13/2022]
Abstract
PURPOSE To describe the experience of our Division of Endocrinology with multikinase inhibitor (MKI) treatment in radioiodine-resistant differentiated thyroid cancer (DTC) patients. METHODS Adults patients with a diagnosis of DTC treated with an MKI drug from March 2011 to October 2018 were registered into a retrospective database. Primary objectives were: the assessment of progression-free survival (PFS) and radiographic response evaluated according to RECIST v. 1.1. Adverse events (AEs) were evaluated by using Common Terminology Criteria for Adverse Events v. 5.0. RESULTS Twenty-two patients were treated with MKIs (21 with sorafenib, one with lenvatinib as first-line treatment). Seven patients required a second-line therapy with lenvatinib and one patient required a third-line treatment with pazopanib. Median duration of treatment was 11.2 (4.8-79.6) months. Best responses with sorafenib were partial response (PR) in two patients (11%), stable disease (SD) >6 months in 13 patients (72%), and progressive disease (PD) in three patients (17%). Best responses with second-line lenvatinib were PR in one patient (33%) and SD in two patients (66%). Median PFS was 31.5 months. AEs were present in 19 (90%) patients under sorafenib. The most common AEs were hand-foot syndrome (HFS) (67%), diarrhea (52%), and hypertension (52%). Definitive withdrawal was necessary in only one patient (4.7%). CONCLUSIONS Our study reflects the real-world clinical experience of an Endocrinology Division on the management of radioiodine-resistant DTC patients with sorafenib and lenvatinib, showing a beneficial therapeutic effect with acceptable tolerability.
Collapse
Affiliation(s)
- Fernando Jerkovich
- Division of Endocrinology, University of Buenos Aires, Buenos, Aires, Argentina
| | | | - Fabián Pitoia
- Division of Endocrinology, University of Buenos Aires, Buenos, Aires, Argentina.
| |
Collapse
|
75
|
Corrigan KL, Williamson H, Elliott Range D, Niedzwiecki D, Brizel DM, Mowery YM. Treatment Outcomes in Anaplastic Thyroid Cancer. J Thyroid Res 2019; 2019:8218949. [PMID: 31249658 PMCID: PMC6556325 DOI: 10.1155/2019/8218949] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Revised: 04/19/2019] [Accepted: 05/09/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Anaplastic thyroid cancer (ATC) is rare, accounting for 1-2% of thyroid malignancies. Median survival is only 3-10 months, and the optimal therapeutic approach has not been established. This study aimed to evaluate outcomes in ATC based on treatment modality. METHODS Retrospective review was performed for patients treated at a single institution between 1990 and 2015. Demographic and clinical covariates were extracted from the medical record. Overall survival (OS) was modeled using Kaplan Meier curves for different treatment modalities. Univariate and multivariate analyses were conducted to assess the relationships between treatment and disease characteristics and OS. RESULTS 28 patients with ATC were identified (n = 16 female, n = 12 male; n = 22 Caucasian, n = 6 African-American; median age 70.9). Majority presented as Stage IVB (71.4%). Most patients received multimodality therapy. 19 patients underwent local surgical resection. 21 patients received locoregional external beam radiotherapy (EBRT) with a median cumulative dose of 3,000 cGy and median number of fractions of 16. 14 patients received systemic therapy (n = 11 concurrent with EBRT), most commonly doxorubicin (n = 9). 16 patients were never disease free, 11 patients had disease recurrence, and 1 patient had no evidence of disease progression. Median OS was 4 months with 1-year survival of 17.9%. Regression analysis showed that EBRT (HR: 0.174; 95% CI: 0.050-0.613; p=0.007) and surgical resection (HR: 0.198; 95% CI: 0.065-0.598; p=0.004) were associated with improved OS. Administration of chemotherapy was not associated with OS. CONCLUSIONS Anaplastic thyroid cancer patients receiving EBRT to the thyroid area/neck and/or surgical resection had better OS than patients without these therapies, though selection bias likely contributed to improved outcomes since patients who can undergo these therapies tend to have better performance status. Prognosis remains poor overall, and new therapeutic approaches are needed to improve outcomes.
Collapse
Affiliation(s)
| | - Hannah Williamson
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, USA
| | | | - Donna Niedzwiecki
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, USA
- Department of Radiation Oncology, Duke Cancer Institute, USA
| | - David M. Brizel
- Duke University School of Medicine, USA
- Department of Radiation Oncology, Duke Cancer Institute, USA
- Department of Surgery, Duke University Medical Center, USA
| | - Yvonne M. Mowery
- Duke University School of Medicine, USA
- Department of Radiation Oncology, Duke Cancer Institute, USA
| |
Collapse
|
76
|
Suri A, Bailey AW, Tavares MT, Gunosewoyo H, Dyer CP, Grupenmacher AT, Piper DR, Horton RA, Tomita T, Kozikowski AP, Roy SM, Sredni ST. Evaluation of Protein Kinase Inhibitors with PLK4 Cross-Over Potential in a Pre-Clinical Model of Cancer. Int J Mol Sci 2019; 20:E2112. [PMID: 31035676 PMCID: PMC6540285 DOI: 10.3390/ijms20092112] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 04/26/2019] [Accepted: 04/26/2019] [Indexed: 12/16/2022] Open
Abstract
Polo-like kinase 4 (PLK4) is a cell cycle-regulated protein kinase (PK) recruited at the centrosome in dividing cells. Its overexpression triggers centrosome amplification, which is associated with genetic instability and carcinogenesis. In previous work, we established that PLK4 is overexpressed in pediatric embryonal brain tumors (EBT). We also demonstrated that PLK4 inhibition exerted a cytostatic effect in EBT cells. Here, we examined an array of PK inhibitors (CFI-400945, CFI-400437, centrinone, centrinone-B, R-1530, axitinib, KW-2449, and alisertib) for their potential crossover to PLK4 by comparative structural docking and activity inhibition in multiple established embryonal tumor cell lines (MON, BT-12, BT-16, DAOY, D283). Our analyses demonstrated that: (1) CFI-400437 had the greatest impact overall, but similar to CFI-400945, it is not optimal for brain exposure. Also, their phenotypic anti-cancer impact may, in part, be a consequence of the inhibition of Aurora kinases (AURKs). (2) Centrinone and centrinone B are the most selective PLK4 inhibitors but they are the least likely to penetrate the brain. (3) KW-2449, R-1530 and axitinib are the ones predicted to have moderate-to-good brain penetration. In conclusion, a new selective PLK4 inhibitor with favorable physiochemical properties for optimal brain exposure can be beneficial for the treatment of EBT.
Collapse
Affiliation(s)
- Amreena Suri
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA.
- Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute, Chicago, IL 60614, USA.
| | - Anders W Bailey
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA.
- Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute, Chicago, IL 60614, USA.
| | - Maurício T Tavares
- Department of Pharmacy, University of São Paulo, São Paulo, SP 05508-900, Brazil.
| | - Hendra Gunosewoyo
- School of Pharmacy and Biomedical Sciences, Curtin University, Bentley, Perth, WA 6102, Australia.
| | - Connor P Dyer
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA.
- Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute, Chicago, IL 60614, USA.
| | - Alex T Grupenmacher
- Department of Ophtalmology, Universidade Federal de São Paulo, São Paulo, SP 04023-062, Brazil.
| | - David R Piper
- Thermo Fisher Scientific, Research and Development, Biosciences Division, Carlsbad, CA 92008, USA.
| | - Robert A Horton
- Thermo Fisher Scientific, Research and Development, Biosciences Division, Carlsbad, CA 92008, USA.
| | - Tadanori Tomita
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA.
- Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute, Chicago, IL 60614, USA.
- Department of Surgery, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA.
| | | | - Saktimayee M Roy
- Department of Pharmacology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA.
| | - Simone T Sredni
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA.
- Cancer Biology and Epigenomics Program, Stanley Manne Children's Research Institute, Chicago, IL 60614, USA.
- Department of Surgery, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
77
|
Shi S, Peng F, Zheng Q, Zeng L, Chen H, Li X, Huang J. Micelle-solubilized axitinib for ocular administration in anti-neovascularization. Int J Pharm 2019; 560:19-26. [PMID: 30710659 DOI: 10.1016/j.ijpharm.2019.01.051] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 12/18/2018] [Accepted: 01/19/2019] [Indexed: 01/24/2023]
Abstract
The development of new blood vessels is directly related to the occurrence of eye diseases. Anti-angiogenic drugs can theoretically be extended to the treatment of ophthalmic diseases. In this study, axitinib, a class of tyrosine kinase inhibitors, was loaded via the amphiphilic copolymer MPEG-PCL, improving its dispersibility in water. Axitinib-loaded micelles showed low toxicity in concentration gradient assays. Additionally, multiple doses by scratch assay confirmed that axitinib had no significant effect on normal cell migration, and biosafety test results showed good cell compatibility. After we established the corneal neovascularization model after an alkali burn in rats, the anti-angiogenic efficacy was tested, with dexamethasone as a positive control. The results showed that axitinib-loaded micelles had anti-angiogenic effects without obvious tissue toxicity. As a class of targeted tyrosine kinase inhibitors, axitinib can be used in the treatment of ocular neovascular diseases through nanocrystallization.
Collapse
Affiliation(s)
- Shuai Shi
- Institute of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou 325027, China.
| | - Fangli Peng
- Institute of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou 325027, China
| | - Qianqian Zheng
- Institute of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou 325027, China
| | - Li Zeng
- Institute of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou 325027, China
| | - Hao Chen
- Institute of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou 325027, China
| | - Xingyi Li
- Institute of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou 325027, China
| | - Jinhai Huang
- Institute of Biomedical Engineering, School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, 270 Xueyuan Road, Wenzhou 325027, China.
| |
Collapse
|
78
|
Ferrari SM, Fallahi P, La Motta C, Elia G, Ragusa F, Ruffilli I, Patrizio A, Baldini E, Ulisse S, Antonelli A. Recent advances in precision medicine for the treatment of anaplastic thyroid cancer. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2019. [DOI: 10.1080/23808993.2019.1565940] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
| | - Poupak Fallahi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | - Giusy Elia
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesca Ragusa
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Ilaria Ruffilli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Armando Patrizio
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Enke Baldini
- Department of Surgical Sciences, ‘Sapienza’ University of Rome, Rome, Italy
| | - Salvatore Ulisse
- Department of Surgical Sciences, ‘Sapienza’ University of Rome, Rome, Italy
| | - Alessandro Antonelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
79
|
Jayarangaiah A, Sidhu G, Brown J, Barrett-Campbell O, Bahtiyar G, Youssef I, Arora S, Skwiersky S, McFarlane SI. Therapeutic options for advanced thyroid cancer. INTERNATIONAL JOURNAL OF CLINICAL ENDOCRINOLOGY AND METABOLISM 2019; 5:26-34. [PMID: 31709422 PMCID: PMC6839707 DOI: 10.17352/ijcem.000040] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Thyroid cancer can be largely classified as well-differentiated, poorly differentiated, medullary and anaplastic. Differentiated thyroid cancer (DTC) includes follicular and papillary subtypes, with the incidence of papillary thyroid cancer (PTC) on the rise. The mainstay of treatment for DTC includes a combination of surgery, radioactive iodine (RAI) and levothyroxine suppression. DTC portends a favorable prognosis, even in the presence of distant metastases, with a 50% rate of 5-year survival largely due to tumor cell's sensitivity to RAI therapy influencing disease outcome. In radioactive iodine refractory differentiated thyroid cancer (RAI-refractory DTC) there is a lower survival rate prompting the use of other therapeutic options available. RAI refractoriness is more common in older patients (age >40), large metastases and lesions that are fluorodeoxyglucose (FDG) avid on position emission tomography (PET). Over the past decade, Identification of genetic mutations in the signaling pathway involved in thyroid tumorigenesis has led to the approval of tyrosine kinase inhibitors (TKIs); Sorafenib and Lenvatinib in RAI-refractory DTC. Similarly, metastatic medullary thyroid cancer (MTC) implies an unfavorable 10-year survival rate of only 20% as the principal treatment options focuses on loco regional control via surgical and/or non-surgical options. The approval of TKIs such as Cabozantinib and Vandetanib has introduced an encouraging, novel, systemic therapeutic option for metastatic MTC. Lastly, anaplastic thyroid cancer (ATC) carries the worst prognosis with high recurrence rates. Treatment includes surgery, chemotherapy and external beam radiation. The FDA recently approved Dabrafenib plus trametinib for BRAF V600E mutated ATC. Considering the modality of chemotherapy and the expanding field of targeted therapies, the role of the oncologist and interaction with endocrinologist in the management of thyroid cancer needs further clarification aiming at collaborative management plans more than ever. This review summarizes the key phase III trials that led to the approval of TKIs in the treatment of DTC and metastatic MTC. Additionally, the review aims to clarify the patient selection criteria for initiation of TKIs and examine the implications, considerations and adverse effects prior to utilizing targeted therapy. Clinical trials are ongoing with promising results and may contribute to the addition of several targeted molecules and immune check point inhibitors to the therapeutic armamentarium for RAI-refractory DTC, medullary and anaplastic thyroid cancer.
Collapse
Affiliation(s)
- Apoorva Jayarangaiah
- Department of Internal Medicine, Jacobi Medical Center, Albert Einstein School of Medicine, New York, USA
| | - Gurinder Sidhu
- Divisions of Endocrinology and Hematology-Oncology, Department of Internal Medicine, State University of New York, Downstate Medical Center, Brooklyn, New York, USA
| | - Jordonna Brown
- Divisions of Endocrinology and Hematology-Oncology, Department of Internal Medicine, State University of New York, Downstate Medical Center, Brooklyn, New York, USA
| | - Odeth Barrett-Campbell
- Divisions of Endocrinology and Hematology-Oncology, Department of Internal Medicine, State University of New York, Downstate Medical Center, Brooklyn, New York, USA
| | - Gul Bahtiyar
- Department of Surgery, Kings County Hospital Center, Brooklyn, New York, USA
| | - Irini Youssef
- Department of Surgery, Kings County Hospital Center, Brooklyn, New York, USA
| | - Shalini Arora
- Department of Surgery, Kings County Hospital Center, Brooklyn, New York, USA
| | - Samara Skwiersky
- Divisions of Endocrinology and Hematology-Oncology, Department of Internal Medicine, State University of New York, Downstate Medical Center, Brooklyn, New York, USA
| | - Samy I McFarlane
- Divisions of Endocrinology and Hematology-Oncology, Department of Internal Medicine, State University of New York, Downstate Medical Center, Brooklyn, New York, USA,Corresponding author: Samy I McFarlane, MD, MPH, MBA, FACP, Distinguished Teaching Professor and Associate Dean, Department of Medicine, Division of Endocrinology, Internal Medicine Residency Program Director, State University of New York-Downstate Medical Center, 450 Clarkson Avenue, Box 50, Brooklyn, New York, 11203, USA, Tel: 718-270-3711; Fax: 718-270-6358;
| |
Collapse
|
80
|
Geller JI, Fox E, Turpin B, Goldstein S, Liu X, Minard CG, Kudgus RA, Reid JM, Berg S, Weigel BJ. A study of axitinib, a VEGF receptor tyrosine kinase inhibitor, in children and adolescents with recurrent or refractory solid tumors: A Children's Oncology Group phase 1 and pilot consortium trial (ADVL1315). Cancer 2018; 124:4548-4555. [PMID: 30394521 PMCID: PMC6289772 DOI: 10.1002/cncr.31725] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 07/11/2018] [Accepted: 07/17/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Axitinib is an oral small molecule that inhibits receptor tyrosine kinases vascular endothelial growth factor receptors 1 to 3. A phase 1 and pharmacokinetic (PK) trial evaluating axitinib was conducted in children with refractory solid tumors. METHODS Axitinib was administered orally twice daily in continuous 28-day cycles. Dose levels (2.4 mg/m2 /dose and 3.2 mg/m2 /dose) were evaluated using a rolling 6 design. Serial PKs (cycle 1, days 1 and 8) and exploratory biomarkers were analyzed. RESULTS A total of 19 patients were enrolled; 1 patient was ineligible due to inadequate time having elapsed from prior therapy. The median age of the patients was 13.5 years (range, 5-17 years). Two of 5 patients who were treated at dose level 2 experienced dose-limiting toxicities (palmar-plantar erythryodysesthesia syndrome in 1 patient and intratumoral hemorrhage in 1 patient). Frequent (>20%) grade 1 to 2 toxicities during cycle 1 included anemia, anorexia, fatigue, diarrhea, nausea, and hypertension. Nonhematological toxicities of grade ≥3 in subsequent cycles included hypertension and elevated serum lipase. PK analysis demonstrated variability in axitinib exposure, the median time to peak plasma concentration was 2 hours, and the half-life ranged from 0.7 to 5.2 hours. Exposure and dose were not found to be significantly associated with hypertension. Five patients achieved stable disease for ≤6 cycles as their best response, including patients with malignant peripheral nerve sheath tumor (1 patient), Ewing sarcoma (1 patient), hepatocellular carcinoma (1 patient), and osteosarcoma (2 patients). One patient with alveolar soft part sarcoma achieved a partial response. Kidney injury biomarkers were found to be elevated at baseline; no trends were identified. CONCLUSIONS In children with refractory solid tumors, the maximum tolerated and recommended dose of axitinib appears to be 2.4 mg/m2 /dose, which provides PK exposures similar to those of adults.
Collapse
Affiliation(s)
- James I. Geller
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH
| | - Elizabeth Fox
- Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Brian Turpin
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH
| | - Stuart Goldstein
- Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH
| | | | - Charles G. Minard
- Dan L. Duncan Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX
| | | | - Joel M. Reid
- Department of Oncology Research, Mayo Clinic, Rochester, MN
| | - Stacey Berg
- Dan L. Duncan Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX
| | - Brenda J. Weigel
- Masonic Children’s Hospital, University of Minnesota Medical Center, Minneapolis, MN
| |
Collapse
|
81
|
Lledó Riquelme M, Campos-Mollo E, Fernández-Sánchez L. Topical axitinib is a potent inhibitor of corneal neovascularization. Clin Exp Ophthalmol 2018; 46:1063-1074. [PMID: 29888852 DOI: 10.1111/ceo.13333] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 04/25/2018] [Accepted: 05/16/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND This study evaluated the effects of topically applied axitinib, a tyrosine kinase inhibitor, in an experimental model of corneal neovascularization (CNV). METHODS A total of 48 New Zealand rabbits were used. CNV was induced by placing five silk sutures in the upper cornea of one eye per rabbit. Rabbits were randomized into four groups (12 rabbits each): 0.9% saline (control group), 0.02 mg/mL axitinib, 0.35 mg/mL axitinib and 0.5 mg/mL axitinib groups. All treatments were administered three times daily for 14 days. Photographs were taken using a slit lamp on days 7 and 14. The area of neovascularization was measured in mm2 , as the percentage of total corneal area and as the percentage of corneal surface covered by sutures (SCS). RESULTS On day 14, the CNV area in the control group (31.50 ± 7.47 mm2 ; 115.00 ± 22.55% of the corneal SCS) was larger than that in the 0.02 mg/mL axitinib group (19.20 ± 8.92 mm2 ; 73.89 ± 34.98%), the 0.35 mg/mL axitinib group (8.83 ± 3.92 mm2 ; 31.90 ± 13.59%) and the 0.5 mg/mL axitinib group (5.12 ± 3.97 mm2 ; 18.38 ± 13.65%). Compared with saline, CNV was inhibited 39.04% by 0.02 mg/mL axitinib, 71.96% by 0.35 mg/mL axitinib and 84.73% by 0.5 mg/mL axitinib. CONCLUSION Topical administration of the three axitinib concentrations inhibited CNV in rabbits, blocking both vascular endothelial growth factor and platelet-derived growth factor pathways. Axitinib at 0.5 mg/mL induced profound inhibition of corneal angiogenesis.
Collapse
Affiliation(s)
| | | | - Laura Fernández-Sánchez
- Department of Physiology, Genetics and Microbiology, University of Alicante, Alicante, Spain
| |
Collapse
|
82
|
Sun Y, Du F, Gao M, Ji Q, Li Z, Zhang Y, Guo Z, Wang J, Chen X, Wang J, Chi Y, Tang P. Anlotinib for the Treatment of Patients with Locally Advanced or Metastatic Medullary Thyroid Cancer. Thyroid 2018; 28:1455-1461. [PMID: 30142994 DOI: 10.1089/thy.2018.0022] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND The prognosis of advanced or metastatic medullary thyroid carcinoma (MTC) is poor, and there are few therapeutic options. Anlotinib has previously shown promising antitumor activity on MTC in preclinical models and a Phase I study. This Phase II clinical trial was devised to confirm the antitumor activity of anlotinib in patients with advanced or metastatic MTC. METHODS Patients with unresectable locally advanced or metastatic MTC received once daily oral anlotinib 12 mg, two weeks on/one week off, until disease progression, death, unacceptable toxicity, or withdrawal of consent for any reason. The dose was adjusted on the basis of observed toxicity. The primary endpoint was progression-free survival (PFS). RESULTS Fifty-eight patients received anlotinib treatment. The primary endpoint PFS has not yet been reached at the time of analysis. On the basis of investigator assessments, 56.9% of patients experienced a partial response. PFS rate at 48 weeks was 85.5%. Forty-five patients had a ≥50% decrease in serum calcitonin concentration from baseline. The most common adverse events were hand-foot syndrome, hypertriglyceridemia, cholesterol elevation, fatigue, and proteinuria. CONCLUSIONS Anlotinib demonstrated a durable antitumor activity with a manageable adverse event profile in locally advanced or metastatic MTC.
Collapse
Affiliation(s)
- Yongkun Sun
- 1 Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital , Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Feng Du
- 2 Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), The VIPII Gastrointestinal Cancer Division of Medical Department, Peking University Cancer Hospital and Institute , Beijing, China
| | - Ming Gao
- 3 Department of Thyroid and Neck Oncology, Tianjin Medical University Cancer Institute and Hospital , Tianjin, China
| | - Qinghai Ji
- 4 Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Zhendong Li
- 5 Department of Head and Neck Surgery, Liaoning Cancer Hospital and Institute , Shenyang, China
| | - Yuan Zhang
- 6 Department of Head and Neck Surgery, Jiangsu Cancer Hospital , Nanjing, China
| | - Zhuming Guo
- 7 Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center , Guangzhou, China
| | - Jun Wang
- 8 Department of Thyroid and Breast Surgery, Gansu Provincial Cancer Hospital , Lanzhou, China
| | - Xiangjin Chen
- 9 Department of Head and Neck Surgery, First Affiliated Hospital of Fujian Medical University , Fuzhou, China
| | - Jinwan Wang
- 1 Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital , Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yihebali Chi
- 1 Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital , Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pingzhang Tang
- 1 Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital , Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
83
|
Del Rivero J, Edgerly M, Ward J, Madan RA, Balasubramaniam S, Fojo T, Gramza AW. Phase I/II Trial of Vandetanib and Bortezomib in Adults with Locally Advanced or Metastatic Medullary Thyroid Cancer. Oncologist 2018; 24:16-e14. [PMID: 30297385 PMCID: PMC6324636 DOI: 10.1634/theoncologist.2018-0452] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 07/24/2018] [Indexed: 02/07/2023] Open
Abstract
Lessons Learned. Vandetanib at a dose of 300 mg orally every day plus bortezomib 1.3 mg/m2 intravenously on days 1, 4, 8, and 11 could be administered safely. Assessing outcomes in 17 patients with medullary thyroid cancer, investigators considered the combination to be more difficult to administer than single‐agent vandetanib and that achieving better outcomes was unlikely. Consequently, a planned phase II study was terminated early.
Background. The proto‐oncogene RET (REarranged during Transfection) has a critical role in the pathogenesis of medullary thyroid cancer (MTC). Vandetanib (V), a multitargeted tyrosine kinase inhibitor approved for the treatment of MTC, is thought to inhibit RET in MTC. Supported by preclinical studies demonstrating that bortezomib (B) administration lowered RET mRNA and protein levels, we conducted a phase I study in advanced solid tumors of vandetanib in combination with bortezomib. The goal was to establish an RP2D (recommended phase II dose) for the combination of vandetanib plus bortezomib, a regimen envisioned as a dual strategy for targeting RET in MTC. Methods. Patients with advanced solid tumors were treated with escalating doses of bortezomib or vandetanib to assess the safety and tolerability of daily oral vandetanib and intravenous (IV) bortezomib administered on days 1, 4, 8, and 11 of a 28‐day cycle. Intrapatient dose escalation was allowed. Results. Twenty‐two patients were enrolled and received escalating mg/m2 bortezomib and mg vandetanib (number of patients) at initial doses of 1 and 100 (3), 1.3 and 100 (6), 1.3 and 200 (6), and 1.3 and 300 (7), respectively. Patients received a median of four cycles of bortezomib/vandetanib (range: 1–10), with 13 patients escalating to 1.3/200 and 10 to 1.3/300. G3 toxicities occurring in more than one patient included hypertension (24%), fatigue (19%), thrombocytopenia (10%), diarrhea (10%), and arthralgia (10%). There were no drug‐related G4/5 toxicities. There was one dose‐limiting toxicity, G3 thrombocytopenia, at bortezomib/vandetanib doses of 1.3/200 in cycle 2 that resolved without intervention. Four patients with a diagnosis of MTC (27%) had a partial response (PR). Conclusion. The MTD of the combination was established as bortezomib, 1.3 mg/m2 IV days 1, 4, 8, and 11 with vandetanib 300 mg p.o. daily. RECIST responses were observed in patients with a diagnosis of MTC.
Collapse
Affiliation(s)
- Jaydira Del Rivero
- Medical Oncology Service/Pediatric Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Maureen Edgerly
- Medical Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Jean Ward
- Murtha Cancer Center, Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Ravi A Madan
- Genitourinary Malignancies, Branch Center for Cancer Research, National Cancer Institute, Bethesda, Maryland, USA
| | | | - Tito Fojo
- Department of Medicine, Division of Hematology Oncology, Columbia University Medical Center, New York New York, USA
- James J. Peters VA Medical Center, Bronx, New York, USA
| | - Ann W Gramza
- Medstar Georgetown Lombardi Comprehensive Cancer Center, Washington, DC, USA
| |
Collapse
|
84
|
Rao SN, Cabanillas ME. Navigating Systemic Therapy in Advanced Thyroid Carcinoma: From Standard of Care to Personalized Therapy and Beyond. J Endocr Soc 2018; 2:1109-1130. [PMID: 30250937 PMCID: PMC6141902 DOI: 10.1210/js.2018-00180] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/08/2018] [Indexed: 12/13/2022] Open
Abstract
Thyroid cancer, with the exception of anaplastic thyroid cancer, typically has very favorable outcomes with the standard therapy. However, those that persist, recur, or metastasize are associated with a worse prognosis. Targeted therapy with kinase inhibitors has shown promise in advanced cases of thyroid cancer, and currently five drug regimens are approved for use in clinical practice in the treatment of differentiated, medullary, and anaplastic thyroid cancer, with more options in the pipeline. However, one of the greatest dilemmas is when and how to initiate one of these drugs, and this is discussed herein.
Collapse
Affiliation(s)
- Sarika N Rao
- MedStar Washington Hospital Center, Washington, DC
- Georgetown University School of Medicine, Washington, DC
| | | |
Collapse
|
85
|
Tasoulas J, Tsourouflis G, Theocharis S. Neovascularization: an attractive but tricky target in thyroid cancer. Expert Opin Ther Targets 2018; 22:799-810. [DOI: 10.1080/14728222.2018.1513494] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Jason Tasoulas
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Gerasimos Tsourouflis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Stamatios Theocharis
- First Department of Pathology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
86
|
Rajabi S, Hedayati M. Medullary Thyroid Cancer: Clinical Characteristics and New Insights into Therapeutic Strategies Targeting Tyrosine Kinases. Mol Diagn Ther 2018; 21:607-620. [PMID: 28698976 DOI: 10.1007/s40291-017-0289-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Medullary thyroid carcinoma (MTC) is a hyperplasia of thyroid C-cells, accounting for 5-10% of all thyroid cancers. MTCs may appear as sporadic or hereditary forms, and several molecules and signaling pathways have been found to function defectively in MTC cells. Tyrosine kinases are the most well-studied molecules that have abnormal function in these tumor cells. Due to their limited response, chemotherapeutic agents and radiation therapy are not effective in treating patients with advanced metastatic MTC. In the past decade, significant attention has been given to the utilization of multikinase inhibitors as targeted therapeutic agents for treating MTC patients, with the most promising results arising from the study of tyrosine kinase inhibitors, which generally bind to the ATP binding sites of these kinases. Two drugs-vandetanib and cabozantinib-are approved for the treatment of aggressive advanced MTC; however, the potential for toxicities and adverse effects of these agents on patient quality of life need to be considered against any therapeutic gain. According to recent data, it appears that inhibition of only one receptor or molecule in a pathway is not as effective as simultaneous inhibition of different pathways, indicating the need to use combination therapy. The main purpose of this review is to describe the clinical characteristics, molecular mechanisms, and current molecular and targeted therapeutic strategies active in clinical trials for advanced MTC treatment.
Collapse
Affiliation(s)
- Sadegh Rajabi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
87
|
Liu JW, Chen C, Loh EW, Chu CC, Wang MY, Ouyang HJ, Chang YT, Zhuang WZ, Chou CW, Huang DJ, Lee CH, Yen Y, Tam KW. Tyrosine kinase inhibitors for advanced or metastatic thyroid cancer: a meta-analysis of randomized controlled trials. Curr Med Res Opin 2018; 34:795-803. [PMID: 28812918 DOI: 10.1080/03007995.2017.1368466] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND AND AIMS Radioiodine-refractory advanced or metastatic thyroid cancer has poor prognosis. We conducted a meta-analysis of randomized controlled trials to evaluate the effectiveness and safety of tyrosine kinase inhibitors (TKIs) for advanced or metastatic thyroid cancer treatment. METHODS Studies published up to April 2017 were selected. The pooled effect size was calculated through meta-analysis by using random effects models. Outcomes were overall survival (OS), progression-free survival (PFS), objective response rate (RR), and adverse events (AEs). RESULTS Six studies examining 1615 patients were included. TKI treatment significantly improved PFS in patients with differentiated thyroid cancer (DTC; hazard ratio [HR] = 0.43; 95% confidence interval [CI], 0.23-0.82) and those with medullary thyroid cancer (MTC; HR = 0.36; 95% CI, 0.22-0.58). TKI treatment significantly prolonged OS in patients with DTC (HR = 0.74; 95% CI, 0.58-0.95). The TKI treatment group exhibited a significantly improved partial response rate (risk ratio = 15.8; 95% CI, 1.77-140.69) but a significantly higher number of AEs compared with the control group. CONCLUSION TKIs significantly improved PFS and RR in patients with advanced or metastatic DTC or MTC. We recommend thoroughly evaluating patients' health status and cautiously using TKIs to maximize their benefits and minimize their toxicity.
Collapse
Affiliation(s)
- Jen-Wei Liu
- a Department of Pharmacy , Shin Kong Wu Ho-Su Memorial Hospital , Taipei , Taiwan
- b School of Pharmacy , College of Pharmacy, Taipei Medical University , Taipei , Taiwan
| | - Chiehfeng Chen
- c Cochrane Taiwan , Taipei Medical University , Taipei , Taiwan
- d Department of Public Health , School of Medicine, College of Medicine, Taipei Medical University , Taipei , Taiwan
- e Division of Plastic Surgery, Department of Surgery , Wan Fang Hospital, Taipei Medical University , Taipei , Taiwan
| | - El-Wui Loh
- f Center for Evidence-Based Health Care , Shuang Ho Hospital, Taipei Medical University , New Taipei City , Taiwan
| | - Chun-Cheng Chu
- g School of Medicine , College of Medicine, Taipei Medical University , Taipei City , Taiwan
| | - Mu-Yi Wang
- g School of Medicine , College of Medicine, Taipei Medical University , Taipei City , Taiwan
| | - Hsin-Ju Ouyang
- g School of Medicine , College of Medicine, Taipei Medical University , Taipei City , Taiwan
| | - Ya-Ting Chang
- g School of Medicine , College of Medicine, Taipei Medical University , Taipei City , Taiwan
| | - Wei-Zhan Zhuang
- g School of Medicine , College of Medicine, Taipei Medical University , Taipei City , Taiwan
| | - Ching-Wen Chou
- g School of Medicine , College of Medicine, Taipei Medical University , Taipei City , Taiwan
| | - Der-Jr Huang
- g School of Medicine , College of Medicine, Taipei Medical University , Taipei City , Taiwan
| | - Chia-Hwa Lee
- h School of Medical Laboratory Science and Biotechnology , College of Medical Science and Technology, Taipei Medical University , Taipei , Taiwan
- i Comprehensive Cancer Center of Taipei Medical University , Taipei , Taiwan
- j Department of Laboratory Medicine , Shuang Ho Hospital, Taipei Medical University , Taipei , Taiwan
| | - Yun Yen
- k Graduate Institute of Cancer Biology and Drug Discovery , College of Medical Science and Technology, Taipei Medical University , Taipei , Taiwan
| | - Ka-Wai Tam
- c Cochrane Taiwan , Taipei Medical University , Taipei , Taiwan
- l Department of Surgery , School of Medicine, College of Medicine, Taipei Medical University , Taipei , Taiwan
| |
Collapse
|
88
|
|
89
|
Berdelou A, Lamartina L, Klain M, Leboulleux S, Schlumberger M. Treatment of refractory thyroid cancer. Endocr Relat Cancer 2018; 25:R209-R223. [PMID: 29371330 DOI: 10.1530/erc-17-0542] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 01/25/2018] [Indexed: 12/22/2022]
Abstract
Distant metastases from thyroid cancer of follicular origin are uncommon. Treatment includes levothyroxine administration, focal treatment modalities with surgery, external radiation therapy and thermal ablation, and radioiodine in patients with uptake of 131I in their metastases. Two-thirds of distant metastases become refractory to radioiodine at some point, and when there is a significant tumor burden and documented progression on imaging, a treatment with a kinase inhibitor may provide benefits.
Collapse
Affiliation(s)
- Amandine Berdelou
- Department of Nuclear Medicine and Endocrine Oncology, TUTHYREF (Tumeurs de la Thyroïde Réfractaires) Network, Gustave Roussy and University Paris-Saclay, Villejuif, France
| | - Livia Lamartina
- Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Michele Klain
- Department of Advanced Biomedical Sciences, University of Naples 'Federico II', Naples, Italy
| | - Sophie Leboulleux
- Department of Nuclear Medicine and Endocrine Oncology, TUTHYREF (Tumeurs de la Thyroïde Réfractaires) Network, Gustave Roussy and University Paris-Saclay, Villejuif, France
| | - Martin Schlumberger
- Department of Nuclear Medicine and Endocrine Oncology, TUTHYREF (Tumeurs de la Thyroïde Réfractaires) Network, Gustave Roussy and University Paris-Saclay, Villejuif, France
| |
Collapse
|
90
|
Gild ML, Topliss DJ, Learoyd D, Parnis F, Tie J, Hughes B, Walsh JP, McLeod DSA, Clifton-Bligh RJ, Robinson BG. Clinical guidance for radioiodine refractory differentiated thyroid cancer. Clin Endocrinol (Oxf) 2018; 88:529-537. [PMID: 29095527 DOI: 10.1111/cen.13508] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 10/25/2017] [Accepted: 10/27/2017] [Indexed: 12/26/2022]
Abstract
Prognosis from differentiated thyroid cancer is worse when the disease becomes refractory to radioiodine. Until recently, treatment options have been limited to local therapies such as surgery and radiotherapy, but the recent availability of systemic therapies now provides some potential for disease control. Multitargeted kinase inhibitors (TKIs) including lenvatinib and sorafenib have been shown to improve progression-free survival in phase III clinical trials, but are also associated with a spectrum of adverse effects. Other TKIs have been utilized as "redifferentiation" agents, increasing sodium iodide symporter expression in metastases and thus restoring radioiodine avidity. Some patients whose disease progresses on initial TKI therapy will still respond to a different TKI and clinical trials currently in progress will clarify the best options for such patients. As these drugs are not inexpensive, care needs to be taken to minimize not only biological but also financial toxicity. In this review, we examine the basic biology of radioiodine refractory disease and discuss optimal treatment approaches, with specific focus on choice and timing of TKI treatment. This clinical field remains fluid, and directions for future research include exploring biomarkers and considering adjuvant TKI use in certain patient groups.
Collapse
Affiliation(s)
- Matti L Gild
- Department of Diabetes, Endocrinology & Metabolism, Royal North Shore Hospital Sydney, St Leonards, NSW, Australia
- The University of Sydney, Sydney, NSW, Australia
| | - Duncan J Topliss
- Department of Endocrinology and Diabetes, The Alfred, Melbourne, VIC, Australia
- Department of Medicine, Monash University, Melbourne, VIC, Australia
| | - Diana Learoyd
- Department of Diabetes, Endocrinology & Metabolism, Royal North Shore Hospital Sydney, St Leonards, NSW, Australia
- The University of Sydney, Sydney, NSW, Australia
| | - Francis Parnis
- Department of Oncology, Adelaide Cancer Centre, Kurralta Park, SA, Australia
- Adelaide University, Adelaide, SA, Australia
| | - Jeanne Tie
- Division of Systems Biology and Personalized Medicine, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
- Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, Australia
- Department of Medical Oncology, Western Health, Melbourne, VIC, Australia
| | - Brett Hughes
- Cancer Care Services, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
- School of Medicine, The University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - John P Walsh
- Department of Endocrinology and Diabetes, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
- School of Medicine and Pharmacology, The University of Western Australia, Crawley, WA, Australia
| | - Donald S A McLeod
- Department of Endocrinology, Diabetes Royal Brisbane and Women's Hospital, Herston, QLD, Australia
- Population Health Department QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Roderick J Clifton-Bligh
- Department of Diabetes, Endocrinology & Metabolism, Royal North Shore Hospital Sydney, St Leonards, NSW, Australia
- The University of Sydney, Sydney, NSW, Australia
- Cancer Genetics Laboratory, Hormones and Cancer Group, Kolling Institute of Medical Research, Sydney, SW, Australia
| | - Bruce G Robinson
- Department of Diabetes, Endocrinology & Metabolism, Royal North Shore Hospital Sydney, St Leonards, NSW, Australia
- The University of Sydney, Sydney, NSW, Australia
- Cancer Genetics Laboratory, Hormones and Cancer Group, Kolling Institute of Medical Research, Sydney, SW, Australia
| |
Collapse
|
91
|
Sugino K, Nagahama M, Kitagawa W, Ohkuwa K, Uruno T, Matsuzu K, Suzuki A, Masaki C, Akaishi J, Hames KY, Tomoda C, Ogimi Y, Ito K. Clinical factors related to the efficacy of tyrosine kinase inhibitor therapy in radioactive iodine refractory recurrent differentiated thyroid cancer patients. Endocr J 2018; 65:299-306. [PMID: 29269689 DOI: 10.1507/endocrj.ej17-0365] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
New insights in thyroid cancer biology propelled the development of targeted therapies as salvage treatment for radioiodine-refractory differentiated thyroid cancer (RR-DTC), and the tyrosine kinase inhibitor (TKI) lenvatinib has recently become available as a new line of therapy for RR-DTC. The aim of this study is to investigate clinical factors related to the efficacy of TKI therapy in recurrent RR-DTC patients and identify the optimal timing for the start of TKI therapy. The subjects consisted of 29 patients with progressive RR-DTC, 9 males and 20 females, median age 66 years. A univariate analysis was conducted in relation to progression free survival (PFS) and overall survival (OS) by the Kaplan-Meier method for the following variables: age, sex, histology of the primary tumor, thyroglobulin doubling time before the start of lenvatinib therapy, site of the target lesions, presence of a tumor-mediated symptom at the start of lenvatinib therapy, and baseline tumor size of the target lesions. Median duration of lenvatinib therapy was 14.7 months and median drug intensity was 9.5 mg. At the time of the data cut-off for the analysis, 9 patients (31.0%) have died of their disease (DOD), and a PR (partial response), SD (stable disease), and PD (progressive disease) were observed in 20 patients (69%), 6 patients (20.7%), 3 patients (10.3%), respectively. Univariate analyses showed that the presence of a symptom was the only factor significantly related to poorer PFS and OS. Clinical benefit of TKI therapy will be possibly limited when the therapy starts after tumor-mediated symptoms appear.
Collapse
Affiliation(s)
| | | | | | - Keiko Ohkuwa
- Department of Surgery, Ito Hospital, Tokyo, Japan
| | | | | | | | - Chie Masaki
- Department of Surgery, Ito Hospital, Tokyo, Japan
| | | | | | | | - Yuna Ogimi
- Department of Surgery, Ito Hospital, Tokyo, Japan
| | - Koichi Ito
- Department of Surgery, Ito Hospital, Tokyo, Japan
| |
Collapse
|
92
|
Abstract
Metastatic renal cell carcinoma (mRCC) is an incurable malignancy, characterized by its resistance to traditional chemotherapy, radiation, and hormonal therapy. Treatment perspectives and prognosis of patients with mRCC have been significantly improved by advances in the understanding of its molecular pathogenesis, which have led to the development of targeted therapeutics. Different molecular factors derived from the tumor or the host detected in both tissue or serum could be predictive of therapeutic benefit. Some of them suggest a rational selection of patients to be treated with certain therapies, though none have been validated for routine use. This article provides an overview of both clinical and molecular factors associated with predictive or prognostic value in mRCC and emphasizes that both should be considered in parallel to provide the most appropriate, individualized treatment and achieve the best outcomes in clinical practice.
Collapse
|
93
|
Jin Y, Van Nostrand D, Cheng L, Liu M, Chen L. Radioiodine refractory differentiated thyroid cancer. Crit Rev Oncol Hematol 2018; 125:111-120. [PMID: 29650270 DOI: 10.1016/j.critrevonc.2018.03.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Revised: 01/22/2018] [Accepted: 03/21/2018] [Indexed: 12/17/2022] Open
Abstract
Differentiated thyroid cancer (DTC) is usually curable with surgery, radioactive iodine (RAI), and thyroid-stimulating hormone (TSH) suppression. However, local recurrence and/or distant metastases occur in approximately 15% of cases during follow-up, and nearly two-thirds of these patients will become RAI-refractory (RR-DTC) with a poor prognosis. This review focuses on the most challenging and rapidly evolving aspects of RR-DTC, and we discuss the considerable improvement in more accurately defining RR-DTC, more effective therapeutic strategies, and describe the diagnosis, pathogenesis, and future prospects of RR-DTC. Along with the detection of serum thyroglobulin and anatomic imaging modalities, such as ultrasound and computer tomography, radionuclide molecular imaging plays a vital role in the evaluation of RR-DTC. In addition, continual progress has been made in the management of RR-DTC, including watchful waiting under appropriate TSH suppression, local treatment approaches, and systemic therapies (molecular targeted therapy, redifferentiation therapy, gene therapy, and cancer immunotherapy). These all hold promise to change the natural history of RR-DTC.
Collapse
Affiliation(s)
- Yuchen Jin
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China.
| | - Douglas Van Nostrand
- MedStar Health Research Institute and Washington Hospital Center, Washington, DC, 20010, United States.
| | - Lingxiao Cheng
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China.
| | - Min Liu
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China.
| | - Libo Chen
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China.
| |
Collapse
|
94
|
Naoum GE, Morkos M, Kim B, Arafat W. Novel targeted therapies and immunotherapy for advanced thyroid cancers. Mol Cancer 2018; 17:51. [PMID: 29455653 PMCID: PMC5817719 DOI: 10.1186/s12943-018-0786-0] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 02/01/2018] [Indexed: 02/06/2023] Open
Abstract
Thyroid cancer is a frequently encountered endocrine malignancy. Despite the favorable prognosis of this disease, 15–20% of differentiated thyroid cancer (DTC) cases and most anaplastic types, remain resistant to standard treatment options, including radioactive iodine (RAI). In addition, around 30% of medullary thyroid cancer (MTC) cases show resistance after surgery. The evolving understanding of disease-specific molecular therapeutic targets has led to the approval of two targeted therapies (Sorafenib and Lenvatinib) for RAI refractory DTC and another two drugs (Vandetanib and Cabozantinib) for MTC. These advanced therapies exert their effects by blocking the MAPK pathway, which has been widely correlated to different types of thyroid cancers. While these drugs remain reserved for thyroid cancer patients who failed all treatment options, their ability to improve patients’ overall survival remain hindered by their low efficacy and other molecular factors. Among these factors is the tumor’s ability to activate parallel proliferative signaling pathways other than the cascades blocked by these drugs, along with overexpression of some tyrosine kinase receptors (TKR). These facts urge the search for novel different treatment strategies for advanced thyroid cases beyond these drugs. Furthermore, the growing knowledge of the dynamic immune system interaction with tumor microenvironment has revolutionized the cancer immune therapy field. In this review, we aim to discuss the molecular escape mechanisms of thyroid tumors from these drugs. We also highlight novel therapeutic options targeting other pathways than MAPK, including PI3K pathway, ALK translocations and HER2/3 receptors and their clinical impact. We also aim to discuss the usage of targeted therapy in restoring thyroid tumor sensitivity to RAI, and finally turn to extensively discuss the role of immunotherapy as a potential alternative treatment option for advanced thyroid diseases.
Collapse
Affiliation(s)
- George E Naoum
- Department of Radiation Oncology, Harvard Medical School, Massachusetts General Hospital, 55 Fruit St, Boston, MA, 02114, USA.,Alexandria Comprehensive Cancer center, Alexandria, Egypt
| | - Michael Morkos
- Department of Endocrinology, Rush University, 1900 W Polk St, Room 801, Chicago, IL, USA
| | - Brian Kim
- Department of Endocrinology, Thyroid Cancer Program, Rush University, Jelke Building, Room 604, 1735 W Harrison St, Chicago, IL, 60612, UK
| | - Waleed Arafat
- Alexandria Comprehensive Cancer center, Alexandria, Egypt. .,University Of Alexandria, Clinical oncology department, Alexandria, Egypt. .,Department of Radiation Oncology, University of Alabama at Birmingham, 1720 2nd Ave S, Birmingham, AL, 35294, UK.
| |
Collapse
|
95
|
Grávalos C, Carrato A, Tobeña M, Rodriguez-Garrote M, Soler G, Vieitez JM, Robles L, Valladares-Ayerbes M, Polo E, Limón ML, Safont MJ, Martínez de Castro E, García-Alfonso P, Aranda E. A Randomized Phase II Study of Axitinib as Maintenance Therapy After First-line Treatment for Metastatic Colorectal Cancer. Clin Colorectal Cancer 2018; 17:e323-e329. [PMID: 29551560 DOI: 10.1016/j.clcc.2018.02.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/12/2018] [Indexed: 12/16/2022]
Abstract
INTRODUCTION The aim of this study was to evaluate the efficacy and safety of maintenance therapy with axitinib versus placebo following induction therapy in patients with metastatic colorectal cancer (mCRC). PATIENTS AND METHODS In this double-blinded, phase II trial, patients with mCRC who had not progressed after 6 to 8 months of first-line chemotherapy were randomized to receive axitinib (5 mg twice a day) (arm A) or placebo (arm B). RESULTS Forty-nine patients were included: 25 in arm A and 24 in arm B. The median follow-up was 26.07 months (95% confidence interval [CI], 18.44-31.73 months). Progression-free survival (PFS) rate at 6 months was 40.00% (95% CI, 21.28%-58.12%) in the axitinib arm versus 8.33% (95% CI, 1.44%-23.30%) in the placebo arm (P = .0141). The median PFS was statistically significantly longer in the axitinib group than in the placebo group (4.96 vs. 3.16 months; hazard ratio, 0.46; 95% CI, 0.25-0.86; P = .0116). Median overall survival was also longer in the axitinib arm but did not reach statistical significance (27.61 vs. 19.99 months; hazard ratio, 0.68; 95% CI, 0.31-1.48; P = .3279). Grade 3 to 4 treatment-related toxicities were experienced by 7 patients (28%) in cohort A and 1 patient (4%) in cohort B (P = .0488). The most frequent grade 3 to 4 treatment-related toxicities were hypertension, diarrhea, and asthenia. There were no toxic deaths. The study was prematurely closed because of slow recruitment. CONCLUSIONS In our study, maintenance treatment with axitinib monotherapy showed a significant increase in PFS and a good safety profile. Axitinib should be further explored as a possible option for first-line chemotherapy maintenance treatment in patients with mCRC.
Collapse
Affiliation(s)
- Cristina Grávalos
- Department of Medical Oncology, University Hospital 12 De Octubre, Madrid, Spain.
| | - Alfredo Carrato
- Department of Medical Oncology, Ramon y Cajal University Hospital, IRYCIS CIBERONC, Madrid, Spain
| | - María Tobeña
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Gemma Soler
- Department of Medical Oncology, ICO Hospital Duran i Reynals, Hospitalet, Spain
| | - José Mª Vieitez
- Department of Medical Oncology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | - Luis Robles
- Department of Medical Oncology, University Hospital 12 De Octubre, Madrid, Spain
| | | | - Eduardo Polo
- Department of Medical Oncology, Hospital Miguel Servet, Zaragoza, Spain
| | - Mª Luisa Limón
- Department of Medical Oncology, Hospital Universitario Virgen del Rocio, Sevilla, Spain
| | - Mª José Safont
- Department of Medical Oncology, Hospital General Universitario Valencia, Valencia, Spain
| | - Eva Martínez de Castro
- Department of Medical Oncology, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Pilar García-Alfonso
- Department of Medical Oncology, Hospital General Universitario Gregorio Marañon, Madrid, Spain
| | - Enrique Aranda
- Department of Medical Oncology, IMIBIC. University Hospital Reina Sofia, CIBERONC Instituto de Salud Carlos III, Cordoba, Spain
| | | |
Collapse
|
96
|
Wells SA. Advances in the management of MEN2: from improved surgical and medical treatment to novel kinase inhibitors. Endocr Relat Cancer 2018; 25:T1-T13. [PMID: 29142004 PMCID: PMC5776732 DOI: 10.1530/erc-17-0325] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 11/06/2017] [Indexed: 12/11/2022]
Abstract
Medullary thyroid carcinoma (MTC), a tumor derived from the neural crest, occurs either sporadically or as the dominant component of the type 2 multiple endocrine neoplasia (MEN) syndromes, MEN2A and MEN2B. The discovery that mutations in the RET protooncogene cause hereditary MTC was of great importance, since it led to the development of novel methods of diagnosis and treatment. For example, the detection of a mutated RET allele in family members at risk for inheriting MEN2A or MEN2B signaled that they would develop MTC, and possibly other components of the syndromes. Furthermore, the detection of a mutated allele created the opportunity, especially in young children, to remove the thyroid before MTC developed, or while it was confined to the gland. The discovery also led to the development of molecular targeted therapeutics (MTTs), mainly tyrosine kinase inhibitors, which were effective in the treatment of patients with locally advanced or metastatic MTC. While responses to MTTs are often dramatic, they are highly variable, and almost always transient, because the tumor cells become resistant to the drugs. Clinical investigators and the pharmaceutical industry are focusing on the development of the next generation of MTTs, which have minimal toxicity and greater specificity for mutated RET.
Collapse
Affiliation(s)
- Samuel A Wells
- Genetics BranchNational Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
97
|
Faugeras L, Pirson AS, Donckier J, Michel L, Lemaire J, Vandervorst S, D'Hondt L. Refractory thyroid carcinoma: which systemic treatment to use? Ther Adv Med Oncol 2018; 10:1758834017752853. [PMID: 29399055 PMCID: PMC5788129 DOI: 10.1177/1758834017752853] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 12/01/2017] [Indexed: 12/21/2022] Open
Abstract
The incidence of thyroid cancer has increased markedly in recent decades, but has been stable in terms of mortality rates. For the most part, these cancers are treated with surgery, which may or may not be followed by radioactive iodine depending on the tumor subtype. Still, many of these cancers will recur and may be treated with radioactive iodine or another surgery. It is unclear what treatment is best for cases of locally advanced or metastatic thyroid cancer that are refractory to radioactive iodine. Chemotherapy has a very low response rate. However, in the past few years, several systemic therapies, primarily targeted, have emerged to improve the overall survival of these patients. Alternative treatments are also of interest, namely peptide receptor radionuclide therapy or immunotherapy.
Collapse
Affiliation(s)
- Laurence Faugeras
- Oncology Department, CHU UCL Namur, 1 rue Therasse, 5530 Yvoir, Belgium
| | | | | | - Luc Michel
- Department of General Surgery, CHU UCL Namur, Yvoir, Belgium
| | - Julien Lemaire
- Department of General Surgery, CHU UCL Namur, Yvoir, Belgium
| | | | | |
Collapse
|
98
|
Molinaro E, Romei C, Biagini A, Sabini E, Agate L, Mazzeo S, Materazzi G, Sellari-Franceschini S, Ribechini A, Torregrossa L, Basolo F, Vitti P, Elisei R. Anaplastic thyroid carcinoma: from clinicopathology to genetics and advanced therapies. Nat Rev Endocrinol 2017; 13:644-660. [PMID: 28707679 DOI: 10.1038/nrendo.2017.76] [Citation(s) in RCA: 318] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Anaplastic thyroid carcinoma (ATC) is a rare malignancy, accounting for 1-2% of all thyroid cancers. Although rare, ATC accounts for the majority of deaths from thyroid carcinoma. ATC often originates in a pre-existing thyroid cancer lesion, as suggested by the simultaneous presence of areas of differentiated or poorly differentiated thyroid carcinoma. ATC is characterized by the accumulation of several oncogenic alterations, and studies have shown that an increased number of oncogenic alterations equates to an increased level of dedifferentiation and aggressiveness. The clinical management of ATC requires a multidisciplinary approach; according to recent American Thyroid Association guidelines, surgery, radiotherapy and/or chemotherapy should be considered. In addition to conventional therapies, novel molecular targeted therapies are the most promising emerging treatment modalities. These drugs are often multiple receptor tyrosine kinase inhibitors, several of which have been tested in clinical trials with encouraging results so far. Accordingly, clinical trials are ongoing to evaluate the safety, efficacy and effectiveness of these new agents. This Review describes the updated clinical and pathological features of ATC and provides insight into the molecular biology of this disease. The most recent literature regarding conventional, newly available and future therapies for ATC is also discussed.
Collapse
Affiliation(s)
- Eleonora Molinaro
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| | - Cristina Romei
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| | - Agnese Biagini
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| | - Elena Sabini
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| | - Laura Agate
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| | - Salvatore Mazzeo
- Diagnostic and Interventional Radiology Department of Translational Research and New Technologies in Medicine and Surgery, University Hospital of Pisa
| | - Gabriele Materazzi
- Division of Endocrine Surgery, Department of Surgical Pathology, University Hospital of Pisa
| | | | | | - Liborio Torregrossa
- Department of Surgical, Medical and Molecular Pathology, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Fulvio Basolo
- Department of Surgical, Medical and Molecular Pathology, University Hospital of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Paolo Vitti
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| | - Rossella Elisei
- Endocrine Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa
| |
Collapse
|
99
|
Suneetha A, Donepudi S. HPLC method development and validation for the estimation of Axitinib in rabbit plasma. BRAZ J PHARM SCI 2017. [DOI: 10.1590/s2175-97902017000300012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
100
|
Off-tumor targets compromise antiangiogenic drug sensitivity by inducing kidney erythropoietin production. Proc Natl Acad Sci U S A 2017; 114:E9635-E9644. [PMID: 29078273 DOI: 10.1073/pnas.1703431114] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Anti-VEGF drugs are commonly used for treatment of a variety of cancers in human patients, and they often develop resistance. The mechanisms underlying anti-VEGF resistance in human cancer patients are largely unknown. Here, we show that in mouse tumor models and in human cancer patients, the anti-VEGF drug-induced kidney hypoxia augments circulating levels of erythropoietin (EPO). Gain-of-function studies show that EPO protects tumor vessels from anti-VEGF treatment and compromises its antitumor effects. Loss of function by blocking EPO function using a pharmacological approach markedly increases antitumor activity of anti-VEGF drugs through inhibition of tumor angiogenesis. Similarly, genetic loss-of-function data shows that deletion of EpoR in nonerythroid cells significantly increases antiangiogenic and antitumor effects of anti-VEGF therapy. Finally, in a relatively large cohort study, we show that treatment of human colorectal cancer patients with bevacizumab augments circulating EPO levels. These findings uncover a mechanism of desensitizing antiangiogenic and anticancer effects by kidney-produced EPO. Our work presents conceptual advances of our understanding of mechanisms underlying antiangiogenic drug resistance.
Collapse
|