1
|
Cortas C, Charalambous H. Tyrosine Kinase Inhibitors for Radioactive Iodine Refractory Differentiated Thyroid Cancer. Life (Basel) 2023; 14:22. [PMID: 38255638 PMCID: PMC10817256 DOI: 10.3390/life14010022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/15/2023] [Accepted: 12/17/2023] [Indexed: 01/24/2024] Open
Abstract
Patients with differentiated thyroid cancer usually present with early-stage disease and undergo surgery followed by adjuvant radioactive iodine ablation, resulting in excellent clinical outcomes and prognosis. However, a minority of patients relapse with metastatic disease, and eventually develop radioactive iodine refractory disease (RAIR). In the past there were limited and ineffective options for systemic therapy for RAIR, but over the last ten to fifteen years the emergence of tyrosine kinase inhibitors (TKIs) has provided important new avenues of treatment for these patients, that are the focus of this review. Currently, Lenvatinib and Sorafenib, multitargeted TKIs, represent the standard first-line systemic treatment options for RAIR thyroid carcinoma, while Cabozantinib is the standard second-line treatment option. Furthermore, targeted therapies for patients with specific targetable molecular abnormalities include Latrectinib or Entrectinib for patients with NTRK gene fusions and Selpercatinib or Pralsetinib for patients with RET gene fusions. Dabrafenib plus Trametinib currently only have tumor agnostic approval in the USA for patients with BRAF V600E mutations, including thyroid cancer. Redifferentiation therapy is an area of active research, with promising initial results, while immunotherapy studies with checkpoint inhibitors in combination with tyrosine kinase inhibitors are underway.
Collapse
Affiliation(s)
| | - Haris Charalambous
- Medical Oncology Department, Bank of Cyprus Oncology Centre, Nicosia 2006, Cyprus;
| |
Collapse
|
2
|
Zhang L, Feng Q, Wang J, Tan Z, Li Q, Ge M. Molecular basis and targeted therapy in thyroid cancer: Progress and opportunities. Biochim Biophys Acta Rev Cancer 2023; 1878:188928. [PMID: 37257629 DOI: 10.1016/j.bbcan.2023.188928] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 05/23/2023] [Accepted: 05/23/2023] [Indexed: 06/02/2023]
Abstract
Thyroid cancer (TC) is the most prevalent endocrine malignant tumor. Surgery, chemotherapy, radiotherapy, and radioactive iodine (RAI) therapy are the standard TC treatment modalities. However, recurrence or tumor metastasis remains the main challenge in the management of anaplastic thyroid cancer (ATC) and radioiodine (RAI) radioactive iodine-refractory differentiated thyroid cancer (RR-DTC). Several multi-tyrosine kinase inhibitors (MKIs), or immune checkpoint inhibitors in combination with MKIs, have emerged as novel therapies for controlling the progression of DTC, medullary thyroid cancer (MTC), and ATC. Here, we discuss and summarize the molecular basis of TC, review molecularly targeted therapeutic drugs in clinical research, and explore potentially novel molecular therapeutic targets. We focused on the evaluation of current and recently emerging tyrosine kinase inhibitors approved for systemic therapy for TC, including lenvatinib, sorafenib and cabozantinib in DTC, vandetanib, cabozantinib, and RET-specific inhibitor (selpercatinib and pralsetinib) in MTC, combination dabrafenib with trametinib in ATC. In addition, we also discuss promising treatments that are in clinical trials and may be incorporated into clinical practice in the future, briefly describe the resistance mechanisms of targeted therapies, emphasizing that personalized medicine is critical to the design of second-line therapies.
Collapse
Affiliation(s)
- Lizhuo Zhang
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang 310014, China
| | - Qingqing Feng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, China.
| | - Jiafeng Wang
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang 310014, China
| | - Zhuo Tan
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang 310014, China.
| | - Qinglin Li
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China.
| | - Minghua Ge
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang 310014, China.
| |
Collapse
|
3
|
Hamidi S, Hofmann MC, Iyer PC, Cabanillas ME, Hu MI, Busaidy NL, Dadu R. Review article: new treatments for advanced differentiated thyroid cancers and potential mechanisms of drug resistance. Front Endocrinol (Lausanne) 2023; 14:1176731. [PMID: 37435488 PMCID: PMC10331470 DOI: 10.3389/fendo.2023.1176731] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/05/2023] [Indexed: 07/13/2023] Open
Abstract
The treatment of advanced, radioiodine refractory, differentiated thyroid cancers (RR-DTCs) has undergone major advancements in the last decade, causing a paradigm shift in the management and prognosis of these patients. Better understanding of the molecular drivers of tumorigenesis and access to next generation sequencing of tumors have led to the development and Food and Drug Administration (FDA)-approval of numerous targeted therapies for RR-DTCs, including antiangiogenic multikinase inhibitors, and more recently, fusion-specific kinase inhibitors such as RET inhibitors and NTRK inhibitors. BRAF + MEK inhibitors have also been approved for BRAF-mutated solid tumors and are routinely used in RR-DTCs in many centers. However, none of the currently available treatments are curative, and most patients will ultimately show progression. Current research efforts are therefore focused on identifying resistance mechanisms to tyrosine kinase inhibitors and ways to overcome them. Various novel treatment strategies are under investigation, including immunotherapy, redifferentiation therapy, and second-generation kinase inhibitors. In this review, we will discuss currently available drugs for advanced RR-DTCs, potential mechanisms of drug resistance and future therapeutic avenues.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ramona Dadu
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
4
|
Ragusa F, Ferrari SM, Elia G, Paparo SR, Balestri E, Botrini C, Patrizio A, Mazzi V, Guglielmi G, Foddis R, Spinelli C, Ulisse S, Antonelli A, Fallahi P. Combination Strategies Involving Immune Checkpoint Inhibitors and Tyrosine Kinase or BRAF Inhibitors in Aggressive Thyroid Cancer. Int J Mol Sci 2022; 23:ijms23105731. [PMID: 35628540 PMCID: PMC9144613 DOI: 10.3390/ijms23105731] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/10/2022] [Accepted: 05/18/2022] [Indexed: 02/01/2023] Open
Abstract
Thyroid cancer is the most common (~90%) type of endocrine-system tumor, accounting for 70% of the deaths from endocrine cancers. In the last years, the high-throughput genomics has been able to identify pathways/molecular targets involved in survival and tumor progression. Targeted therapy and immunotherapy individually have many limitations. Regarding the first one, although it greatly reduces the size of the cancer, clinical responses are generally transient and often lead to cancer relapse after initial treatment. For the second one, although it induces longer-lasting responses in cancer patients than targeted therapy, its response rate is lower. The individual limitations of these two different types of therapies can be overcome by combining them. Here, we discuss MAPK pathway inhibitors, i.e., BRAF and MEK inhibitors, combined with checkpoint inhibitors targeting PD-1, PD-L1, and CTLA-4. Several mutations make tumors resistant to treatments. Therefore, more studies are needed to investigate the patient's individual tumor mutation burden in order to overcome the problem of resistance to therapy and to develop new combination therapies.
Collapse
Affiliation(s)
- Francesca Ragusa
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, 56126 Pisa, Italy; (F.R.); (G.E.); (S.R.P.); (E.B.); (C.B.); (V.M.); (C.S.)
| | - Silvia Martina Ferrari
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy;
| | - Giusy Elia
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, 56126 Pisa, Italy; (F.R.); (G.E.); (S.R.P.); (E.B.); (C.B.); (V.M.); (C.S.)
| | - Sabrina Rosaria Paparo
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, 56126 Pisa, Italy; (F.R.); (G.E.); (S.R.P.); (E.B.); (C.B.); (V.M.); (C.S.)
| | - Eugenia Balestri
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, 56126 Pisa, Italy; (F.R.); (G.E.); (S.R.P.); (E.B.); (C.B.); (V.M.); (C.S.)
| | - Chiara Botrini
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, 56126 Pisa, Italy; (F.R.); (G.E.); (S.R.P.); (E.B.); (C.B.); (V.M.); (C.S.)
| | - Armando Patrizio
- Department of Emergency Medicine, Azienda Ospedaliero-Universitaria Pisana, 56124 Pisa, Italy;
| | - Valeria Mazzi
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, 56126 Pisa, Italy; (F.R.); (G.E.); (S.R.P.); (E.B.); (C.B.); (V.M.); (C.S.)
| | - Giovanni Guglielmi
- U.O. Medicina Preventiva Del Lavoro, Azienda Ospedaliero-Universitaria Pisana, 56124 Pisa, Italy;
| | - Rudy Foddis
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (R.F.); (P.F.)
| | - Claudio Spinelli
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, 56126 Pisa, Italy; (F.R.); (G.E.); (S.R.P.); (E.B.); (C.B.); (V.M.); (C.S.)
| | - Salvatore Ulisse
- Department of Surgical Sciences, ‘Sapienza’ University of Rome, 00161 Rome, Italy;
| | - Alessandro Antonelli
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, 56126 Pisa, Italy; (F.R.); (G.E.); (S.R.P.); (E.B.); (C.B.); (V.M.); (C.S.)
- Correspondence: ; Tel.: +39-050-992318
| | - Poupak Fallahi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy; (R.F.); (P.F.)
| |
Collapse
|
5
|
Nacchio M, Pisapia P, Pepe F, Russo G, Vigliar E, Porcelli T, Luongo C, Iaccarino A, Pagni F, Salvatore D, Troncone G, Malapelle U, Bellevicine C. Predictive molecular pathology in metastatic thyroid cancer: the role of RET fusions. Expert Rev Endocrinol Metab 2022; 17:167-178. [PMID: 35404189 DOI: 10.1080/17446651.2022.2060819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 03/29/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Rearranged during transfection (RET) gene fusions are detected in 10-20% of thyroid cancer patients. Recently, RET fusion-positive metastatic thyroid cancers have attracted much attention owing to the FDA approval of two highly selective anti-RET tyrosine kinase inhibitors, namely, selpercatinib, and pralsetinib. AREAS COVERED This review summarizes the available evidence on the biological and predictive role of RET gene fusions in thyroid carcinoma patients and the latest screening assays currently used to detect these genomic alterations in histological and cytological specimens. EXPERT OPINION Management of advanced thyroid carcinoma has significantly evolved over the last decade thanks to the approval of three multikinase inhibitors, i.e. sorafenib, lenvatinib, cabozantinib, and of two selective RET-tyrosine inhibitors, i.e. selpercatinib and pralsetinib. In this setting, the detection of RET-fusions in advanced thyroid cancer specimens through the use of next-generation sequencing has become a commonly used strategy in clinical practice to select the best treatment options.
Collapse
Affiliation(s)
- Mariantonia Nacchio
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Pasquale Pisapia
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Gianluca Russo
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Elena Vigliar
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Tommaso Porcelli
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Cristina Luongo
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Antonino Iaccarino
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, San Gerardo Hospital, University of Milano-Bicocca, Monza, Italy
| | - Domenico Salvatore
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Claudio Bellevicine
- Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
6
|
Pitoia F, Jerkovich F, Trimboli P, Smulever A. New approaches for patients with advanced radioiodine-refractory thyroid cancer. World J Clin Oncol 2022; 13:9-27. [PMID: 35116229 PMCID: PMC8790300 DOI: 10.5306/wjco.v13.i1.9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 08/31/2021] [Accepted: 12/31/2021] [Indexed: 02/06/2023] Open
Abstract
The cumulative evidence over the past decades has shown that the incidence of differentiated thyroid carcinoma (DTC) has exponentially increased. Approximately 10% of patients with DTC exhibit recurrent or metastatic disease, and about two-thirds of the latter will be defined as refractory to radioactive iodine (RAIR) treatment. Since this condition implies 10-year survival rates less than 10% after detection, using available treatments, such as systemic and targeted therapies, have become increasingly relevant. The initiation of these treatments aims to reach stabilization, tumor volume reduction, and/or symptom improvement and it should be decided by highly specialized endocrinologists/oncologists on the basis of patient's features. Considering that despite enlarged progression-free survival was proven, multikinase inhibitors remain non-curative, their benefits last for a limited time and the side effects potentially cause harm and quality of life reduction. In this context, molecular testing of cancer cells provides a promising spectrum of targeted therapies that offer increased compatibility with individual patient needs by improving efficacy, progression free survival, overall survival and adverse events profile. This review article aims to provide a summary of the current therapeutic strategies in advanced RAIR-DTC, including approved target therapies as well as those for off-label use, RAI resensitization agents, and immunotherapy.
Collapse
Affiliation(s)
- Fabián Pitoia
- Division of Endocrinology, Hospital de Clínicas José de San Martin, University of Buenos Aires, Buenos Aires 1120, Argentina
| | - Fernando Jerkovich
- Division of Endocrinology, Hospital de Clínicas José de San Martin, University of Buenos Aires, Buenos Aires 1120, Argentina
| | - Pierpaolo Trimboli
- Clinic for Endocrinology and Diabetology, Lugano Regional Hospital, Ente Ospedaliero Cantonale, Lugano 1111, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano 1111, Switzerland
| | - Anabella Smulever
- Division of Endocrinology, Hospital de Clínicas José de San Martin, University of Buenos Aires, Buenos Aires 1120, Argentina
| |
Collapse
|
7
|
Qu N, Hui Z, Shen Z, Kan C, Hou N, Sun X, Han F. Thyroid Cancer and COVID-19: Prospects for Therapeutic Approaches and Drug Development. Front Endocrinol (Lausanne) 2022; 13:873027. [PMID: 35600591 PMCID: PMC9114699 DOI: 10.3389/fendo.2022.873027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 04/04/2022] [Indexed: 02/05/2023] Open
Abstract
Thyroid cancer is the most prevalent endocrine malignancy and the reported incidence of thyroid cancer has continued to increase in recent years. Since 2019, coronavirus disease 2019 (COVID-19) has been spreading worldwide in a global pandemic. COVID-19 aggravates primary illnesses and affects disease management; relevant changes include delayed diagnosis and treatment. The thyroid is an endocrine organ that is susceptible to autoimmune attack; thus, thyroid cancer after COVID-19 has gradually attracted attention. Whether COVID-19 affects the diagnosis and treatment of thyroid cancer has also attracted the attention of many researchers. This review examines the literature regarding the influence of COVID-19 on the pathogenesis, diagnosis, and treatment of thyroid cancer; it also focuses on drug therapies to promote research into strategies for improving therapy and management in thyroid cancer patients with COVID-19.
Collapse
Affiliation(s)
- Na Qu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Zongguang Hui
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Zhixin Shen
- Department of Breast and Thyroid Surgery, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Chengxia Kan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Ningning Hou
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiaodong Sun
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
- *Correspondence: Fang Han, ; Xiaodong Sun,
| | - Fang Han
- Department of Endocrinology and Metabolism, Affiliated Hospital of Weifang Medical University, Weifang, China
- Clinical Research Center, Affiliated Hospital of Weifang Medical University, Weifang, China
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
- *Correspondence: Fang Han, ; Xiaodong Sun,
| |
Collapse
|
8
|
Puliafito I, Esposito F, Prestifilippo A, Marchisotta S, Sciacca D, Vitale MP, Giuffrida D. Target Therapy in Thyroid Cancer: Current Challenge in Clinical Use of Tyrosine Kinase Inhibitors and Management of Side Effects. Front Endocrinol (Lausanne) 2022; 13:860671. [PMID: 35872981 PMCID: PMC9304687 DOI: 10.3389/fendo.2022.860671] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 05/12/2022] [Indexed: 01/18/2023] Open
Abstract
Thyroid cancer (TC) is the most common endocrine malignancy. TC is classified as differentiated TC (DTC), which includes papillary and follicular subtypes and Hürthle cell variants, medullary TC (MTC), anaplastic TC (ATC), and poorly differentiated TC (PDTC). The standard of care in DTC consists of surgery together with radioactive iodine (131I) therapy and thyroid hormone, but patients with MTC do not benefit from 131I therapy. Patients with advanced TC resistant to 131I treatment (RAI-R) have no chance of cure, as well as patients affected by ATC and progressive MTC, in which conventional therapy plays only a palliative role, representing, until a few years ago, an urgent unmet need. In the last decade, a better understanding of molecular pathways involved in the tumorigenesis of specific histopathological subtypes of TC has led to develop tyrosine kinase inhibitors (TKIs). TKIs represent a valid treatment in progressive advanced disease and were tested in all subtypes of TC, highlighting the need to improve progression-free survival. However, treatments using these novel therapeutics are often accompanied by side effects that required optimal management to minimize their toxicities and thereby enable patients who show benefit to continue treatment and obtain maximal clinical efficacy. The goal of this overview is to provide an update on the current use of the main drugs recently studied for advanced TC and the management of the adverse events.
Collapse
Affiliation(s)
- Ivana Puliafito
- Medical Oncology Unit, Istituto Oncologico del Mediterraneo SpA, Viagrande, Italy
| | - Francesca Esposito
- IOM Ricerca Srl, Viagrande, Italy
- *Correspondence: Francesca Esposito, ; Dario Giuffrida,
| | - Angela Prestifilippo
- Medical Oncology Unit, Istituto Oncologico del Mediterraneo SpA, Viagrande, Italy
| | | | - Dorotea Sciacca
- Medical Oncology Unit, Istituto Oncologico del Mediterraneo SpA, Viagrande, Italy
| | - Maria Paola Vitale
- Hospital Pharmacy Unit, Istituto Oncologico del Mediterraneo SpA, Viagrande, Italy
| | - Dario Giuffrida
- Medical Oncology Unit, Istituto Oncologico del Mediterraneo SpA, Viagrande, Italy
- *Correspondence: Francesca Esposito, ; Dario Giuffrida,
| |
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW We review the new systemic treatment strategies for differentiated thyroid carcinoma, as well as the acquaintance of its molecular biology. RECENT FINDINGS Multiple kinase inhibitor drugs have become the standard therapy for thyroid cancer, albeit several adverse effects. In the last few years, new molecules have raised with an overall safety profile. Most of them, are considered targeted therapies directed toward driven-molecules alterations, such as neurotrophic tyrosine kinase receptor (NTRK) inhibitors for NTRK-fusion thyroid cancer and rearranged during transfection (RET) inhibitors for RET-fusion thyroid cancer. Recently, promising outcomes and safety data have been presented. Furthermore, other novel strategies for advanced thyroid carcinoma are currently investigated in clinical trials.The ability to provide precision medicine to patients in routine clinical settings depends on the availability of molecular profiling test at their cancer centers. The impossibility to perform molecular characterization could turn out to be a diagnostic and treatment limitation for some patients. SUMMARY The treatment of advanced differentiated thyroid carcinoma has undergone rapid evolution in the last decade. An emerging treatment era is coming. From now to then, we will need to face the different types of diagnostic tools for molecular characterization, their interpretation and, finally the access to targeted therapies.
Collapse
|
10
|
Lin YS, Zhang X, Wang C, Liu YQ, Guan WM, Liang J. Long-Term Results of a Phase II Trial of Apatinib for Progressive Radioiodine Refractory Differentiated Thyroid Cancer. J Clin Endocrinol Metab 2021; 106:e3027-e3036. [PMID: 33769497 DOI: 10.1210/clinem/dgab196] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Indexed: 12/13/2022]
Abstract
CONTEXT Radioiodine refractory differentiated thyroid cancer (RAIR-DTC) has been a global challenge due to its poor prognosis and limited treatment options. OBJECTIVE We report here the long-term results of the phase II clinical trial of apatinib, an anti-angiogenic tyrosine kinase inhibitor, for RAIR-DTC. METHODS This was an open-label, exploratory phase II clinical trial among progressive RAIR-DTC patients. Apatinib treatment was given once daily until disease progression, unmanageable toxicity, withdrawal, or death. The primary end points were objective response rate (ORR) and disease control rate (DCR). Progression-free survival (PFS), overall survival (OS), duration of response, long-term safety, and the association between patients with different tumor genotype (BRAFV600E and TERT promotor mutation) and their PFS rates were also assessed. RESULTS The ORR was 80%, and the DCR was 95%. The overall median PFS was 18.4 months (95% CI, 9.2-36.8 months) and the median OS was 51.6 months (95% CI, 29.2-not reached [NR]). Patients with BRAFV600E mutation (10 of 18 evaluated) had a longer median PFS compared with patients with BRAF wild-type (NR vs 9.2 months; P = 0.002). The most common adverse events included palmar-plantar erythrodysesthesia syndrome (19/20), proteinuria (18/20), and hypertension (16/20). CONCLUSION In this long-term evaluation, apatinib displayed sustainable efficacy and tolerable safety profile, warranting it as a promising treatment option for progressive RAIR-DTC.
Collapse
Affiliation(s)
- Yan-Song Lin
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Beijing, Chinese Academy of Medical Sciences & PUMC, 100730, Beijing, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, 100730, Beijing, China
| | - Xin Zhang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Beijing, Chinese Academy of Medical Sciences & PUMC, 100730, Beijing, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, 100730, Beijing, China
| | - Chen Wang
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Beijing, Chinese Academy of Medical Sciences & PUMC, 100730, Beijing, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, 100730, Beijing, China
| | - Yan-Qing Liu
- Department of Nuclear Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College (PUMC) Hospital, Beijing, Chinese Academy of Medical Sciences & PUMC, 100730, Beijing, China
- Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, 100730, Beijing, China
| | - Wen-Min Guan
- Department of Radiology, Peking Union Medical College (PUMC) Hospital, Beijing, Chinese Academy of Medical Sciences & PUMC, Beijing, 100730, China
| | - Jun Liang
- Department of Oncology, Peking University International Hospital, Beijing, 102206, China
| |
Collapse
|
11
|
Melaccio A, Sgaramella LI, Pasculli A, Di Meo G, Gurrado A, Prete FP, Vacca A, Ria R, Testini M. Prognostic and Therapeutic Role of Angiogenic Microenvironment in Thyroid Cancer. Cancers (Basel) 2021; 13:cancers13112775. [PMID: 34204889 PMCID: PMC8199761 DOI: 10.3390/cancers13112775] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/29/2021] [Accepted: 05/31/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Angiogenesis is an essential event for the progression of solid tumors and is promoted by angiogenic cytokines released in the tumor microenvironment by neoplastic and stromal cells. Over the last 20 years, the role of the microenvironment and the implication of several angiogenic factors in tumorigenesis of solid and hematological neoplasms have been widely studied. The tumor microenvironment has also been well-defined for thyroid cancer, clarifying the importance of angiogenesis in cancer progression, spread, and metastasis. Furthermore, recent studies have evaluated the association of circulating angiogenic factors with the clinical outcomes of differentiated thyroid cancer, potentially providing noninvasive, low-cost, and safe tests that can be used in screening, diagnosis, and follow-up. In this review, we highlight the mechanisms of action of these proangiogenic factors and their different molecular pathways, as well as their applications in the treatment and prognosis of thyroid cancer. Abstract Thyroid cancer is the most common endocrine malignancy, with a typically favorable prognosis following standard treatments, such as surgical resection and radioiodine therapy. A subset of thyroid cancers progress to refractory/metastatic disease. Understanding how the tumor microenvironment is transformed into an angiogenic microenvironment has a role of primary importance in the aggressive behavior of these neoplasms. During tumor growth and progression, angiogenesis represents a deregulated biological process, and the angiogenic switch, characterized by the formation of new vessels, induces tumor cell proliferation, local invasion, and hematogenous metastases. This evidence has propelled the scientific community’s effort to study a number of molecular pathways (proliferation, cell cycle control, and angiogenic processes), identifying mediators that may represent viable targets for new anticancer treatments. Herein, we sought to review angiogenesis in thyroid cancer and the potential role of proangiogenic cytokines for risk stratification of patients. We also present the current status of treatment of advanced differentiated, medullary, and poorly differentiated thyroid cancers with multiple tyrosine kinase inhibitors, based on the rationale of angiogenesis as a potential therapeutic target.
Collapse
Affiliation(s)
- Assunta Melaccio
- Operative Unit of Internal Medicine “G. Baccelli”, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro” Medical School, 70124 Bari, Italy; (A.M.); (A.V.); (R.R.)
| | - Lucia Ilaria Sgaramella
- Academic General Surgery Unit “V. Bonomo”, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro” Medical School, 70124 Bari, Italy; (L.I.S.); (A.P.); (G.D.M.); (A.G.); (F.P.P.)
| | - Alessandro Pasculli
- Academic General Surgery Unit “V. Bonomo”, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro” Medical School, 70124 Bari, Italy; (L.I.S.); (A.P.); (G.D.M.); (A.G.); (F.P.P.)
| | - Giovanna Di Meo
- Academic General Surgery Unit “V. Bonomo”, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro” Medical School, 70124 Bari, Italy; (L.I.S.); (A.P.); (G.D.M.); (A.G.); (F.P.P.)
| | - Angela Gurrado
- Academic General Surgery Unit “V. Bonomo”, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro” Medical School, 70124 Bari, Italy; (L.I.S.); (A.P.); (G.D.M.); (A.G.); (F.P.P.)
| | - Francesco Paolo Prete
- Academic General Surgery Unit “V. Bonomo”, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro” Medical School, 70124 Bari, Italy; (L.I.S.); (A.P.); (G.D.M.); (A.G.); (F.P.P.)
| | - Angelo Vacca
- Operative Unit of Internal Medicine “G. Baccelli”, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro” Medical School, 70124 Bari, Italy; (A.M.); (A.V.); (R.R.)
| | - Roberto Ria
- Operative Unit of Internal Medicine “G. Baccelli”, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro” Medical School, 70124 Bari, Italy; (A.M.); (A.V.); (R.R.)
| | - Mario Testini
- Academic General Surgery Unit “V. Bonomo”, Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro” Medical School, 70124 Bari, Italy; (L.I.S.); (A.P.); (G.D.M.); (A.G.); (F.P.P.)
- Correspondence: ; Tel.: +39-3355370914
| |
Collapse
|
12
|
Marotta V, Chiofalo MG, Di Gennaro F, Daponte A, Sandomenico F, Vallone P, Costigliola L, Botti G, Ionna F, Pezzullo L. Kinase-inhibitors for iodine-refractory differentiated thyroid cancer: still far from a structured therapeutic algorithm. Crit Rev Oncol Hematol 2021; 162:103353. [PMID: 34000414 DOI: 10.1016/j.critrevonc.2021.103353] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 01/16/2021] [Accepted: 05/11/2021] [Indexed: 12/17/2022] Open
Abstract
The kinase-inhibitors (KIs) sorafenib and lenvatinib demonstrated efficacy in iodine-refractory DTC upon phase III studies. However, evidence allowing a punctual balance of benefits and risks is poor. Furthermore, the lack of a direct comparison hampers to establish the proper sequence of administration. However, some insights may provided: a) indirect comparison between phase III trials showed milder toxicity for sorafenib, which should be preferred in case of cardiovascular comorbidities; b) prospective evidence of efficacy in KIs pre-treated patients is available only for lenvatinib, which should be used as second-line. Promising activity was found for the majority of other tested KIs, but no placebo-controlled trials are available. Emerging, but still early, frontiers include the restoration of iodine-sensitivity and the selective activity on pathogenic mutations. In conclusion, the use of KIs in iodine-refractory DTC is far from a structured therapeutic algorithm.
Collapse
Affiliation(s)
- Vincenzo Marotta
- Struttura Complessa Chirurgia Oncologica Della Tiroide, Istituto Nazionale Tumori - Irccs - Fondazione G.Pascale, Napoli, Italy.
| | - Maria Grazia Chiofalo
- Struttura Complessa Chirurgia Oncologica Della Tiroide, Istituto Nazionale Tumori - Irccs - Fondazione G.Pascale, Napoli, Italy
| | - Francesca Di Gennaro
- Struttura Complessa Medicina Nucleare e Terapia Metabolica, Istituto Nazionale Tumori - Irccs - Fondazione G.Pascale, Napoli, Italy
| | - Antonio Daponte
- Struttura Complessa Oncologia Clinica Sperimentale Testa-Collo e Muscolo-Scheletrica, Istituto Nazionale Tumori - Irccs - Fondazione G.Pascale, Napoli, Italy
| | - Fabio Sandomenico
- Struttura Complessa Radiodiagnostica, Istituto Nazionale Tumori - Irccs - Fondazione G.Pascale, Napoli, Italy
| | - Paolo Vallone
- Struttura Complessa Radiodiagnostica, Istituto Nazionale Tumori - Irccs - Fondazione G.Pascale, Napoli, Italy
| | - Luciana Costigliola
- Unità Operativa Compessa Di Chirugia Generale, d'Urgenza e Metabolica, Pineta Grande Hospital, Castel Volturno, Italy
| | - Gerardo Botti
- Struttura Complessa Anatomia Patologica e Citopatologia, Istituto Nazionale Tumori - Irccs - Fondazione G.Pascale, Napoli, Italy
| | - Franco Ionna
- Struttura Complessa Chirurgia Oncologica Maxillo-Facciale Ed ORL, Istituto Nazionale Tumori - Irccs - Fondazione G.Pascale, Napoli, Italy
| | - Luciano Pezzullo
- Struttura Complessa Chirurgia Oncologica Della Tiroide, Istituto Nazionale Tumori - Irccs - Fondazione G.Pascale, Napoli, Italy.
| |
Collapse
|
13
|
Efstathiadou ZA, Tsentidis C, Bargiota A, Daraki V, Kotsa K, Ntali G, Papanastasiou L, Tigas S, Toulis K, Pazaitou-Panayiotou K, Alevizaki M. Benefits and Limitations of TKIs in Patients with Medullary Thyroid Cancer: A Systematic Review and Meta-Analysis. Eur Thyroid J 2021; 10:125-139. [PMID: 33981617 PMCID: PMC8077374 DOI: 10.1159/000509457] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/16/2020] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Tyrosine kinase inhibitors (TKIs) have been used in patients with advanced medullary thyroid carcinoma (MTC); however, data on their effectiveness and safety are limited. The aim of this systematic review and meta-analysis was to document clinical response and toxicities of TKIs in advanced MTC. METHODS We systematically searched major databases for articles or abstracts on TKI use in MTC patients until May 2018. Objective response (OR), defined as the sum of complete + partial response, expressed as percentage, was our primary endpoint, while disease stability, disease progression (DP), median progression-free survival (PFS), and drug discontinuation rate due to adverse events (AEs) were secondary endpoints. Pooled percentages, PFS time, and 95% CIs were reported. RESULTS Thirty-three publications were finally included in the analysis: 1 phase IV, 2 phase III trials evaluating vandetanib and cabozantinib, respectively, 20 phase I or II studies, and the remaining 10 studies of retrospective-observational nature. OR was documented in 28.6% (95% CI 25.9-31.9) of patients. Stable disease was recorded in 46.2% (95% CI 43.3-49.1). Overall, DP was observed in 22.9% (95% CI 20.4-27.6). Grade 3 or more AEs occurred in 48.5% (95% CI 45.5-51.5) of patients, and drug discontinuation was reported in 44.7% (95% CI 41.7-47.6). In general, use of TKIs conferred a PFS of 23.3 months (95% CI 21.07-25.5). In particular, vandetanib induced an OR in 33.8% (95% CI 29.6-38.0) of patients and cabozantinib in 27.7% (95% CI 22.05-33.4). DP occurred in 23.7% (95% CI 19.9-27.6) with vandetanib use and in 22.6% (95% CI 17.4-27.9) in cabozantinib-treated patients. Sorafenib, the third most frequently studied drug, showed intermediate efficacy, but higher discontinuation rates. CONCLUSION Treatment with TKIs in MTC patients with progressive disease is associated with a moderate therapeutic benefit, with achievement of either disease stability or partial response in 73%. The toxicity of these drugs is not negligible, but it is, nonetheless, manageable.
Collapse
Affiliation(s)
- Zoe A. Efstathiadou
- Department of Endocrinology, “Hippokration” General Hospital of Thessaloniki, Thessaloniki, Greece
- *Zoe A. Efstathiadou, Department of Endocrinology, “Hippokration” General Hospital of Thessaloniki, Konstantinoupoleos 49, GR–54642 Thessaloniki (Greece),
| | - Charalambos Tsentidis
- Department of Endocrinology, General Hospital of Nikaia “Agios Panteleimon”, Piraeus, Greece
| | | | - Vasiliki Daraki
- Department of Endocrinology, University Hospital of Crete, Heraklion, Greece
| | - Kalliopi Kotsa
- Department of Endocrinology, “Ahepa” Hospital, Aristotle University, Thessaloniki, Greece
| | - Georgia Ntali
- Department of Endocrinology, Diabetes and Metabolism, “Evangelismos” Hospital Athens, Athens, Greece
| | - Labrini Papanastasiou
- Department of Endocrinology and Diabetes Center, Athens General Hospital “G. Gennimatas”, Athens, Greece
| | - Stelios Tigas
- Department of Endocrinology, University of Ioannina, Ioannina, Greece
| | | | | | - Maria Alevizaki
- Endocrine Unit, Department of Medical Therapeutics, School of Medicine, Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
14
|
Liu G, Chen T, Ding Z, Wang Y, Wei Y, Wei X. Inhibition of FGF-FGFR and VEGF-VEGFR signalling in cancer treatment. Cell Prolif 2021; 54:e13009. [PMID: 33655556 PMCID: PMC8016646 DOI: 10.1111/cpr.13009] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 01/18/2021] [Accepted: 01/29/2021] [Indexed: 02/05/2023] Open
Abstract
The sites of targeted therapy are limited and need to be expanded. The FGF‐FGFR signalling plays pivotal roles in the oncogenic process, and FGF/FGFR inhibitors are a promising method to treat FGFR‐altered tumours. The VEGF‐VEGFR signalling is the most crucial pathway to induce angiogenesis, and inhibiting this cascade has already got success in treating tumours. While both their efficacy and antitumour spectrum are limited, combining FGF/FGFR inhibitors with VEGF/VEGFR inhibitors are an excellent way to optimize the curative effect and expand the antitumour range because their combination can target both tumour cells and the tumour microenvironment. In addition, biomarkers need to be developed to predict the efficacy, and combination with immune checkpoint inhibitors is a promising direction in the future. The article will discuss the FGF‐FGFR signalling pathway, the VEGF‐VEGFR signalling pathway, the rationale of combining these two signalling pathways and recent small‐molecule FGFR/VEGFR inhibitors based on clinical trials.
Collapse
Affiliation(s)
- Guihong Liu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Chen
- Cardiology Department, Chengdu NO.7 People's Hospital, Chengdu Tumor Hospital, Chengdu, China
| | - Zhenyu Ding
- Department of Biotherapy, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Wang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Abstract
BACKGROUND Genomic aberrations (mutations, gene fusions, amplifications) and dysregulation of the fibroblast growth factor (FGF) receptor (FGFR) signaling pathway are frequently found in squamous cell carcinomas of the head and neck (HNSCCs). Targeted therapy with tyrosine kinase inhibitors (TKIs) or monoclonal antibodies directed against FGF receptors therefore represents a promising approach for the treatment of HNSCC. OBJECTIVE This review article describes the current status of FGFR-directed therapies for head and neck tumors (especially HNSCC) and, in this context, discusses genomic alterations of the FGFR pathway as potential companion predictive biomarkers. METHODS This article is based on searches of PubMed, ClinicalTrials.gov, and conference proceedings. RESULTS First results prove the efficacy of TKIs both in HNSCC and in adenocarcinomas of the head and neck, especially in thyroid and adenocystic salivary gland carcinomas. CONCLUSION Early clinical and preclinical data point to the promise of biomarker-directed treatment of patients with head and neck tumors using FGFR-targeted TKIs.
Collapse
Affiliation(s)
- Dimo Dietrich
- Klinik und Poliklinik für Hals-Nasen-Ohrenheilkunde/Chirurgie, Universitätsklinikum Bonn, Venusberg-Campus 1, 53127, Bonn, Deutschland.
| |
Collapse
|
16
|
Belli C, Penault-Llorca F, Ladanyi M, Normanno N, Scoazec JY, Lacroix L, Reis-Filho JS, Subbiah V, Gainor JF, Endris V, Repetto M, Drilon A, Scarpa A, André F, Douillard JY, Curigliano G. ESMO recommendations on the standard methods to detect RET fusions and mutations in daily practice and clinical research. Ann Oncol 2021; 32:337-350. [PMID: 33455880 DOI: 10.1016/j.annonc.2020.11.021] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 11/26/2020] [Accepted: 11/28/2020] [Indexed: 12/17/2022] Open
Abstract
Aberrant activation of RET is a critical driver of growth and proliferation in diverse solid tumours. Multikinase inhibitors (MKIs) showing anti-RET activities have been tested in RET-altered tumours with variable results. The low target specificity with consequent increase in side-effects and off-target toxicities resulting in dose reduction and drug discontinuation are some of the major issues with MKIs. To overcome these issues, new selective RET inhibitors such as pralsetinib (BLU-667) and selpercatinib (LOXO-292) have been developed in clinical trials, with selpercatinib recently approved by the Food and Drug Administration (FDA). The results of these trials showed marked and durable antitumour activity and manageable toxicity profiles in patients with RET-altered tumours. The European Society for Medical Oncology (ESMO) Translational Research and Precision Medicine Working Group (TR and PM WG) launched a collaborative project to review the available methods for the detection of RET gene alterations, their potential applications and strategies for the implementation of a rational approach for the detection of RET fusion genes and mutations in human malignancies. We present here recommendations for the routine clinical detection of targetable RET rearrangements and mutations.
Collapse
Affiliation(s)
- C Belli
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - F Penault-Llorca
- University Clermont Auvergne, INSERM U1240, Centre Jean Perrin, Department of BioPathology, Clermont-Ferrand, France
| | - M Ladanyi
- Department of Pathology and Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, USA
| | - N Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione G. Pascale"-IRCCS, Naples, Italy
| | - J-Y Scoazec
- AMMICa, CNRS-UMS 3655 and INSERM-US23, Gustave Roussy, Villejuif, France; Department of Pathology and Translational Research, Gustave Roussy Cancer Centre, Villejuif, France
| | - L Lacroix
- Translational Research Laboratory and Biobank, Gustave Roussy, Villejuif, France; Inserm U981, Gustave Roussy, Villejuif, France; Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
| | - J S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, USA
| | - V Subbiah
- The University of Texas MD Anderson Cancer Center, Houston, USA
| | - J F Gainor
- Massachusetts General Hospital, Boston, USA
| | - V Endris
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - M Repetto
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - A Drilon
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, USA
| | - A Scarpa
- ARC-Net Research Centre and Department of Diagnostics and Public Health - Section of Pathology, University of Verona, Verona, Italy
| | - F André
- Gustave Roussy Cancer Center, Villejuif, France
| | - J-Y Douillard
- Scientific and Medical Division, European Society for Medical Oncology, Lugano, Switzerland
| | - G Curigliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
17
|
Liu M, Chen P, Hu HY, Ou-Yang DJ, Khushbu RA, Tan HL, Huang P, Chang S. Kinase gene fusions: roles and therapeutic value in progressive and refractory papillary thyroid cancer. J Cancer Res Clin Oncol 2021; 147:323-337. [PMID: 33387037 DOI: 10.1007/s00432-020-03491-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/02/2020] [Indexed: 12/19/2022]
Abstract
The incidence of papillary thyroid cancer (PTC), the major type of thyroid cancer, is increasing rapidly around the world, and its pathogenesis is still unclear. There is poor prognosis for PTC involved in rapidly progressive tumors and resistance to radioiodine therapy. Kinase gene fusions have been discovered to be present in a wide variety of malignant tumors, and an increasing number of novel types have been detected in PTC, especially progressive tumors. As a tumor-driving event, kinase fusions are constitutively activated or overexpress their kinase function, conferring oncogenic potential, and their frequency is second only to BRAFV600E mutation in PTC. Diverse forms of kinase fusions have been observed and are associated with specific pathological features of PTC (usually at an advanced stage), and clinical trials of therapeutic strategies targeting kinase gene fusions are feasible for radioiodine-resistant PTC. This review summarizes the roles of kinase gene fusions in PTC and the value of clinical therapy of targeting fusions in progressive or refractory PTC, and discusses the future perspectives and challenges related to kinase gene fusions in PTC patients.
Collapse
Affiliation(s)
- Mian Liu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Pei Chen
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Hui-Yu Hu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Deng-Jie Ou-Yang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Rooh-Afza Khushbu
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Hai-Long Tan
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Peng Huang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Shi Chang
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410008, China.
| |
Collapse
|
18
|
Ding S, Wang R, Peng S, Luo X, Zhong L, Yang H, Ma Y, Chen S, Wang W. Targeted therapies for RET-fusion cancer: Dilemmas and breakthrough. Biomed Pharmacother 2020; 132:110901. [PMID: 33125973 DOI: 10.1016/j.biopha.2020.110901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 10/04/2020] [Accepted: 10/14/2020] [Indexed: 11/29/2022] Open
Abstract
Genomic profiling has revolutionized treatment options for patients with oncogene-driven cancers, such as epidermal growth factor receptor (EGFR) mutant carcinoma. Rearranged during transfection (RET) rearrangement, as one of the main activated oncogenes, has been well studied and found to be involved in the malignant behavior of carcinogenesis, resulting in acquired resistance to EGFR tyrosine kinase inhibitors and inducing an intrinsic resistance to immunotherapy. Thus, targeted therapies have been investigated against RET arrangement cancers, including several multi-kinase inhibitors and selective RET inhibitors. However, modest efficacy, a relatively high rate of toxicity, and poor effectiveness against brain metastasis are common limitations of multi-targeted novel molecular inhibitors. A promising prospect was shown recently in selective RET inhibitors in several ongoing clinical trials. In this review, we reviewed the concurrent dilemmas of targeted therapies against RET arrangement cancer from preclinical and clinical studies and proposed several clinical considerations for clinical practice prospectively.
Collapse
Affiliation(s)
- SiJie Ding
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Rong Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - ShunLi Peng
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Xiaoqing Luo
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - LongHui Zhong
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Hong Yang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China; Department of Oncology, Hunan Provincial People's Hospital and The First Affiliated Hospital of Hunan Normal University, Changsha, Hunan, PR China
| | - YueYun Ma
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - ShiYu Chen
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China
| | - Wei Wang
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, PR China.
| |
Collapse
|
19
|
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: 1.0] [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
|
20
|
Kachko VA, Platonova NM, Vanushko VE, Shifman BM. [The role of molecular testing in thyroid tumors]. ACTA ACUST UNITED AC 2020; 66:33-46. [PMID: 33351337 DOI: 10.14341/probl12491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 06/14/2020] [Accepted: 06/28/2020] [Indexed: 12/11/2022]
Abstract
¹I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; ²Endocrinology Research Centre, Moscow, Russia Thyroid cancer is the most common endocrine gland cancer. In the last few decades, the molecular diagnostics for thyroid tumors have been widely researched. It is one of the few cancers whose incidence has increased in recent years from microcarcinomas to common, large forms, in all age groups, from children to the elder people. Most researches focus on the genetic basis, since our current knowledge of the genetic background of various forms of thyroid cancer is far from being complete. Molecular and genetic research has several main directions: firstly, differential diagnosis of thyroid tumors, secondly, the prognostic value of detected mutations in thyroid cancer, and thirdly, targeted therapy for aggressive or radioactive iodine-resistant forms of thyroid cancer. In this review, we wanted to update our understanding and describe the prevailing advances in molecular genetics of thyroid cancer, focusing on the main genes associated with the pathology and their potential application in clinical practice.
Collapse
Affiliation(s)
- Vera A Kachko
- I.M. Sechenov First Moscow State Medical University (Sechenov University)
| | | | | | | |
Collapse
|
21
|
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: 49] [Impact Index Per Article: 12.3] [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
|
22
|
Belli C, Anand S, Gainor JF, Penault-Llorca F, Subbiah V, Drilon A, Andrè F, Curigliano G. Progresses Toward Precision Medicine in RET-altered Solid Tumors. Clin Cancer Res 2020; 26:6102-6111. [PMID: 32665298 DOI: 10.1158/1078-0432.ccr-20-1587] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/08/2020] [Accepted: 07/10/2020] [Indexed: 11/16/2022]
Abstract
RET (rearranged during transfection) gene encodes a receptor tyrosine kinase essential for many physiologic functions, but RET aberrations are involved in many pathologies. While RET loss-of-function mutations are associated with congenital disorders like Hirschsprung disease and CAKUT, RET gain-of-function mutations and rearrangements are critical drivers of tumor growth and proliferation in many different cancers. RET-altered (RET+ ) tumors have been hitherto targeted with multikinase inhibitors (MKI) having anti-RET activities, but they inhibit other kinase targets more potently and show limited clinical activities. The lack of target specificity and consequently increased side effects, responsible for dose reduction and drug discontinuation, are critical limitations of MKIs in the clinics. New selective RET inhibitors, selpercatinib and pralsetinib, are showing promising activities, improved response rates, and more favorable toxicity profiles in early clinical trials. This review critically discusses the oncogenic activation of RET and its role in different kinds of tumors, clinical features of RET+ tumors, clinically actionable genetic RET alterations and their diagnosis, and the available data and results of nonselective and selective targeting of RET.
Collapse
Affiliation(s)
- Carmen Belli
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Santosh Anand
- Department of Genetic Medicine and Development (GEDEV), Faculty of Medicine, University of Geneva Medical School, Geneva, Switzerland.,Department of Informatics, Systems and Communications (DISCo), University of Milano-Bicocca, Milan, Italy
| | | | - Frederique Penault-Llorca
- Department of Biopathology, Centre Jean Perrin and University Clermont Auvergne/INSERM U1240, Clermont-Ferrand, France
| | - Vivek Subbiah
- The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexander Drilon
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, New York.,Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, New York
| | | | - Giuseppe Curigliano
- Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy. .,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
23
|
Taylor MH, Alva AS, Larson T, Szpakowski S, Purkaystha D, Amin A, Karpiak L, Piha-Paul SA. A mutation-specific, single-arm, phase 2 study of dovitinib in patients with advanced malignancies. Oncotarget 2020; 11:1235-1243. [PMID: 32292573 PMCID: PMC7147086 DOI: 10.18632/oncotarget.27530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/03/2020] [Indexed: 01/24/2023] Open
Abstract
Background: Receptor tyrosine kinases (RTKs) play key roles in tumorigenesis. The multi-RTK inhibitor dovitinib has demonstrated promising antitumor activity in multiple cancers. Patients and Methods: In this phase 2, open-label, single-arm study, patients with advanced malignancies with RTK-pathway genetic aberrations whose disease progressed on/following standard treatment received dovitinib (500 mg/day; 5-days-on/2-days-off). The primary endpoint was clinical benefit rate (CBR; complete response, partial response [PR], or stable disease [SD] for ≥ 16 weeks). Results: Of 80 patients enrolled, common tumors included gastrointestinal stromal tumors (GIST; 20.0%), colorectal cancer (CRC; 18.8%), and ovarian cancer (10.0%). Patients were heavily pretreated (median prior lines = 4; 67.5% had ≥ 3 prior lines). Genetic aberrations included cKIT (28.8%), FGFR3 (15.0%), and RET (15.0%). The CBR was 13.8%; one PR (GIST) and 10 SD (adenoid cystic [n = 3]; ovarian [n = 3]; GIST [n = 2]; CRC [n = 1]; gastroesophageal junction [n = 1]). The most common treatment-related adverse events were fatigue, diarrhea, nausea, and vomiting. Conclusions: In this heterogeneous patient population, the safety profile was acceptable for dovitinib therapy. A subset of patients with RTK pathway-activated tumors experienced clinical benefit. However, the primary endpoint was not met, suggesting further refinement of predictive biomarkers is required.
Collapse
Affiliation(s)
- Matthew H Taylor
- Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
| | - Ajjai S Alva
- Division of Hematology and Oncology, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Das Purkaystha
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Alpesh Amin
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Linda Karpiak
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Sarina A Piha-Paul
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
24
|
Subbiah V, Yang D, Velcheti V, Drilon A, Meric-Bernstam F. State-of-the-Art Strategies for Targeting RET-Dependent Cancers. J Clin Oncol 2020; 38:1209-1221. [PMID: 32083997 DOI: 10.1200/jco.19.02551] [Citation(s) in RCA: 165] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Activating receptor tyrosine kinase RET (rarranged during transfection) gene alterations have been identified as oncogenic in multiple malignancies. RET gene rearrangements retaining the kinase domain are oncogenic drivers in papillary thyroid cancer, non-small-cell lung cancer, and multiple other cancers. Activating RET mutations are associated with different phenotypes of multiple endocrine neoplasia type 2 as well as sporadic medullary thyroid cancer. RET is thus an attractive therapeutic target in patients with oncogenic RET alterations. Multikinase inhibitors with RET inhibitor activity, such as cabozantinib and vandetanib, have been explored in the clinic for tumors with activating RET gene alterations with modest clinical efficacy. As a result of the nonselective nature of these multikinase inhibitors, patients had off-target adverse effects, such as hypertension, rash, and diarrhea. This resulted in a narrow therapeutic index of these drugs, limiting ability to dose for clinically effective RET inhibition. In contrast, the recent discovery and clinical validation of highly potent selective RET inhibitors (pralsetinib, selpercatinib) demonstrating improved efficacy and a more favorable toxicity profile are poised to alter the landscape of RET-dependent cancers. These drugs appear to have broad activity across tumors with activating RET alterations. The mechanisms of resistance to these next-generation highly selective RET inhibitors is an area of active research. This review summarizes the current understanding of RET alterations and the state-of-the-art treatment strategies in RET-dependent cancers.
Collapse
Affiliation(s)
- Vivek Subbiah
- Department of Investigational Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX.,Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX.,MD Anderson Cancer Network, Houston, TX
| | - Dong Yang
- Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Alexander Drilon
- Thoracic Oncology Service, Early Drug Development Service, Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Department of Medicine, Weill Cornell Medical College, New York, NY
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX.,Sheikh Khalifa Bin Zayed Al Nahyan Institute for Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
25
|
AlRabiah H, Kadi AA, Aljohar HI, Attwa MW, Al-Shakliah NS, Attia SM, Mostafa GA. A New Validated HPLC-MS/MS Method for Quantification and Pharmacokinetic Evaluation of Dovitinib, a Multi-Kinase Inhibitor, in Mouse Plasma. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:407-415. [PMID: 32095071 PMCID: PMC6995292 DOI: 10.2147/dddt.s223573] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 01/03/2020] [Indexed: 12/25/2022]
Abstract
Background Dovitinib (TKI 258) is a small-molecule multi-kinase inhibitor for the treatment of different types of cancer. There is currently no validated method for its quantitative determination; therefore, we aimed to develop a reliable method to assay dovitinib. Method and Results An electrospray ionization tandem mass spectrometry (ESI-MS/MS) method was used to separate dovitinib using an analytical C18 column (50 × 2.1 mm, 1.8 μm) at 25°C. Bosutinib was used as the internal standard (IS). Dovitinib was extracted from mouse plasma using a precipitation procedure. The mobile phase consisted of 10 mM ammonium formate: acetonitrile (68:32, v/v, pH 4.3) run at a rate of 0.3 mL min−1. MS detection was performed in the positive ion mode. Multiple reaction monitoring transitions were 393→337 and 393→309 for dovitinib, and 530→141 and 530→113 for bosutinib. The investigated method was validated as a bio-analytical method based on FDA guidelines. The linearity of the developed method was over the range of 5–500 ng mL,−1 coefficient of determination (r2= 0.9998). The average intra-day recovery and relative standard deviation (RSD) of the quality control (QC) sample were 97.24% and 1.32%, whereas the overall inter-day accuracy and precision were 97.99% and 0.54%, respectively. Dovitinib was stable during sample storage and handling conditions. Furthermore, the dilution integrity of the method was demonstrated by good recovery (97–99%) and RSD values (0.5–0.7%). Conclusion This method was selectively sensitive and exhibited no matrix effect, with an acceptable accuracy and precision according to the FDA guidelines. The developed method could be efficiently used for pharmacokinetic studies of dovitinib.
Collapse
Affiliation(s)
- Haitham AlRabiah
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11459, Saudi Arabia
| | - Adnan A Kadi
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11459, Saudi Arabia
| | - Haya I Aljohar
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11459, Saudi Arabia
| | - Mohamed W Attwa
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11459, Saudi Arabia
| | - Nasser S Al-Shakliah
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11459, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Gamal Ae Mostafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11459, Saudi Arabia.,Micro-Analytical Laboratory, Applied Organic Chemistry Department, National Research Center, Dokki, Cairo, Egypt
| |
Collapse
|
26
|
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: 188] [Impact Index Per Article: 37.6] [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
|
27
|
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: 87] [Impact Index Per Article: 17.4] [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
|
28
|
Brose MS, Bible KC, Chow LQM, Gilbert J, Grande C, Worden F, Haddad R. Management of treatment-related toxicities in advanced medullary thyroid cancer. Cancer Treat Rev 2018; 66:64-73. [PMID: 29704768 DOI: 10.1016/j.ctrv.2018.04.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 04/12/2018] [Accepted: 04/20/2018] [Indexed: 10/17/2022]
Abstract
Progress in the treatment of advanced medullary thyroid cancer (MTC) has resulted from the approval of 2 drugs within the past 5 years, vandetanib and cabozantinib. These multikinase inhibitors (MKIs) possess overlapping specificities for multiple kinase targets implicated in the progression of MTC. Both drugs are associated with toxicities, including hypertension, hemorrhage/perforation, diarrhea and other gastrointestinal events, several dermatologic events, and hypothyroidism. In addition, vandetanib is uniquely associated with QTc prolongation through interaction with myocardial potassium channels, and cabozantinib is uniquely associated with hand-foot skin reaction. Treatment-related toxicities occur frequently and can be severe or life-threatening, and patients undergoing long-term treatment will likely experience adverse events (AEs). Here we offer specific practical recommendations for managing AEs commonly occurring with vandetanib and cabozantinib. The recommended approach relies on early recognition and palliation of symptoms, dose interruption, and dose reduction as necessary in order for the patient to maintain the highest tolerable dose for as long as possible and optimal quality of life. Treatment guidelines do not specify a recommended sequence for treating with vandetanib and cabozantinib; however, most patients will receive both drugs during their lifetime. The choice for first-line therapy is individualized after a risk-benefit assessment and depends on physician preference and patient-related factors, such as comorbid conditions. Because most generalist practices may not be familiar with the intricacies of agents such as vandetanib and cabozantinib, we commend that patients with advanced MTC be managed and treated by a thyroid cancer specialist with coordination of care within a multidisciplinary team.
Collapse
Affiliation(s)
- Marcia S Brose
- Department of Otorhinolaryngology, Head and Neck Surgery and the Abramson Cancer Center, University of Pennsylvania, United States
| | | | | | | | - Carolyn Grande
- Department of Otorhinolaryngology, Head and Neck Surgery and the Abramson Cancer Center, University of Pennsylvania, United States
| | | | | |
Collapse
|
29
|
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: 53] [Impact Index Per Article: 8.8] [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
|
30
|
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: 152] [Impact Index Per Article: 25.3] [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
|
31
|
Targeting RET-driven cancers: lessons from evolving preclinical and clinical landscapes. Nat Rev Clin Oncol 2017; 15:151-167. [PMID: 29134959 DOI: 10.1038/nrclinonc.2017.175] [Citation(s) in RCA: 219] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The gene encoding the receptor-tyrosine kinase RET was first discovered more than three decades ago, and activating RET rearrangements and mutations have since been identified as actionable drivers of oncogenesis. Several multikinase inhibitors with activity against RET have been explored in the clinic, and confirmed responses to targeted therapy with these agents have been observed in patients with RET-rearranged lung cancers or RET-mutant thyroid cancers. Nevertheless, response rates to RET-directed therapy are modest compared with those achieved using targeted therapies matched to other oncogenic drivers of solid tumours, such as sensitizing EGFR or BRAFV600E mutations, or ALK or ROS1 rearrangements. To date, no RET-directed targeted therapeutic has received regulatory approval for the treatment of molecularly defined populations of patients with RET-mutant or RET-rearranged solid tumours. In this Review, we discuss how emerging data have informed the debate over whether the limited success of multikinase inhibitors with activity against RET can be attributed to the tractability of RET as a drug target or to the lack, until 2017, of highly specific inhibitors of this oncoprotein in the clinic. We emphasize that novel approaches to targeting RET-dependent tumours are necessary to improve the clinical efficacy of single-agent multikinase inhibition and, thus, hasten approvals of RET-directed targeted therapies.
Collapse
|
32
|
A drug–drug interaction study to assess the effect of the CYP1A2 inhibitor fluvoxamine on the pharmacokinetics of dovitinib (TKI258) in patients with advanced solid tumors. Cancer Chemother Pharmacol 2017; 81:73-80. [DOI: 10.1007/s00280-017-3469-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 10/19/2017] [Indexed: 01/12/2023]
|
33
|
Thanasupawat T, Natarajan S, Rommel A, Glogowska A, Bergen H, Krcek J, Pitz M, Beiko J, Krawitz S, Verma IM, Ghavami S, Klonisch T, Hombach-Klonisch S. Dovitinib enhances temozolomide efficacy in glioblastoma cells. Mol Oncol 2017; 11:1078-1098. [PMID: 28500786 PMCID: PMC5537714 DOI: 10.1002/1878-0261.12076] [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: 04/19/2017] [Accepted: 04/25/2017] [Indexed: 12/15/2022] Open
Abstract
The multikinase inhibitor and FDA‐approved drug dovitinib (Dov) crosses the blood–brain barrier and was recently used as single drug application in clinical trials for GB patients with recurrent disease. The Dov‐mediated molecular mechanisms in GB cells are unknown. We used GB patient cells and cell lines to show that Dov downregulated the stem cell protein Lin28 and its target high‐mobility group protein A2 (HMGA2). The Dov‐induced reduction in pSTAT3Tyr705 phosphorylation demonstrated that Dov negatively affects the STAT3/LIN28/Let‐7/HMGA2 regulatory axis in GB cells. Consistent with the known function of LIN28 and HMGA2 in GB self‐renewal, Dov reduced GB tumor sphere formation. Dov treatment also caused the downregulation of key base excision repair factors and O6‐methylguanine‐DNA‐methyltransferase (MGMT), which are known to have important roles in the repair of temozolomide (TMZ)‐induced alkylating DNA damage. Combined Dov/TMZ treatment enhanced TMZ‐induced DNA damage as quantified by nuclear γH2AX foci and comet assays, and increased GB cell apoptosis. Pretreatment of GB cells with Dov (‘Dov priming’) prior to TMZ treatment reduced GB cell viability independent of p53 status. Sequential treatment involving ‘Dov priming’ and alternating treatment cycles with TMZ and Dov substantially reduced long‐term GB cell survival in MGMT+ patient GB cells. Our results may have immediate clinical implications to improve TMZ response in patients with LIN28+/HMGA2+GB, independent of their MGMT methylation status.
Collapse
Affiliation(s)
| | - Suchitra Natarajan
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Canada
| | - Amy Rommel
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Aleksandra Glogowska
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Canada
| | - Hugo Bergen
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Canada
| | - Jerry Krcek
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Canada.,Department of Surgery, University of Manitoba, Winnipeg, Canada
| | - Marshall Pitz
- Department of Internal Medicine, University of Manitoba, Winnipeg, Canada
| | - Jason Beiko
- Department of Surgery, University of Manitoba, Winnipeg, Canada
| | - Sherry Krawitz
- Department of Pathology, University of Manitoba, Winnipeg, Canada
| | - Inder M Verma
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Canada
| | - Thomas Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Canada.,Department of Surgery, University of Manitoba, Winnipeg, Canada.,Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Canada
| | - Sabine Hombach-Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Canada.,Obstetrics, Gynecology and Reproductive Medicine, College of Medicine, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
34
|
Lirov R, Worden FP, Cohen MS. The Treatment of Advanced Thyroid Cancer in the Age of Novel Targeted Therapies. Drugs 2017; 77:733-745. [PMID: 28361210 PMCID: PMC5683961 DOI: 10.1007/s40265-017-0733-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Until recently, patients with advanced thyroid cancers had limited options for systemic treatment. With the introduction of tyrosine kinase inhibitors (TKIs) as a promising new class of targeted therapies for thyroid cancer, suddenly patients with advanced disease were given new options to extend survival. Guidelines worldwide have been updated to include general indications for these newer agents, but questions remain regarding which agent(s) to select, when to begin treatment, and how long therapy should continue. Additionally, the true impact of TKIs on overall survival and quality-of-life in thyroid cancer patients needs further clarification. As familiarity with approved agents and longer-term data become available, better strategies for implementation of these targeted drugs will evolve to optimize benefit for patients living with metastatic disease.
Collapse
Affiliation(s)
- Roy Lirov
- Division of Endocrine Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Surgery, University of Michigan Hospital and Health Systems, 2920K Taubman Center, SPC 5331, 1500 East Medical Center Drive, Ann Arbor, MI, 48109-5331, USA
| | - Francis P Worden
- Division of Medical Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Mark S Cohen
- Division of Endocrine Surgery, University of Michigan, Ann Arbor, MI, USA.
- Department of Surgery, University of Michigan Hospital and Health Systems, 2920K Taubman Center, SPC 5331, 1500 East Medical Center Drive, Ann Arbor, MI, 48109-5331, USA.
| |
Collapse
|
35
|
Lorusso L, Pieruzzi L, Biagini A, Sabini E, Valerio L, Giani C, Passannanti P, Pontillo-Contillo B, Battaglia V, Mazzeo S, Molinaro E, Elisei R. Lenvatinib and other tyrosine kinase inhibitors for the treatment of radioiodine refractory, advanced, and progressive thyroid cancer. Onco Targets Ther 2016; 9:6467-6477. [PMID: 27799794 PMCID: PMC5079697 DOI: 10.2147/ott.s84625] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Lenvatinib is a small oral molecule able to inhibit three of the extracellular and intracellular molecules involved in the modulation of angiogenesis and lymphangiogenesis: vascular endothelial growth factor receptor 1–3, fibroblast growth factor receptor 1–4, and platelet-derived growth factor receptor alpha. Since it is also able to inhibit the REarranged during Transfection oncogene and the protooncogene c-KIT, this drug can also be used to control tumor cell proliferation. The maximum tolerated dose, as demonstrated in Phase I studies, is 25 mg daily. The drug is rapidly absorbed with maximum concentrations achieved within 3 and 5 hours after administration in fasting and nonfasting treated patients, respectively. The most common adverse events, reported in Phase I study and confirmed in the subsequent Phase II and III studies, are hypertension, proteinuria, and gastrointestinal symptoms such as nausea, diarrhea, and stomatitis. In Phase I studies, efficacy of lenvatinib in solid tumors was demonstrated, and these encouraging results have led to the development of a Phase II study using lenvatinib in advance radioiodine-refractory differentiated thyroid cancer (DTCs) patients. Since an overall response rate of 50% was reported, this study also confirmed the efficacy of lenvatinib in DTCs patients with an acceptable toxicity profile. Recently, a Phase III study in patients with DTCs (SELECT study) demonstrated the lenvatinib efficacy in prolonging progression-free survival with respect to the placebo (18.3 vs 3.6 months; P<0.001). Although there was no statistically significant difference in the overall survival of the entire group, this result was observed when the analysis was restricted to both the follicular histotype and the group of senior patients (>65 years). The study confirmed that the most common side effects of this drug are hypertension, diarrhea, decreased appetite, weight loss, nausea, and proteinuria. In this review, we report the results of the main studies on lenvatinib efficacy in patients with advanced and progressive thyroid cancer, mainly in DTCs but also in medullary and anaplastic thyroid cancer. We also compared the efficacy of lenvatinib with that of other tyrosine kinase inhibitors, mainly sorafenib, already tested in the same type of patient population.
Collapse
Affiliation(s)
- Loredana Lorusso
- Endocrine Unit, Department of Clinical and Experimental Medicine
| | - Letizia Pieruzzi
- Endocrine Unit, Department of Clinical and Experimental Medicine
| | - Agnese Biagini
- Endocrine Unit, Department of Clinical and Experimental Medicine
| | - Elena Sabini
- Endocrine Unit, Department of Clinical and Experimental Medicine
| | - Laura Valerio
- Endocrine Unit, Department of Clinical and Experimental Medicine
| | - Carlotta Giani
- Endocrine Unit, Department of Clinical and Experimental Medicine
| | | | | | - Valentina Battaglia
- Division of Diagnostic and Interventional Radiology, University of Pisa, Pisa, Italy
| | - Salvatore Mazzeo
- Division of Diagnostic and Interventional Radiology, University of Pisa, Pisa, Italy
| | | | - Rossella Elisei
- Endocrine Unit, Department of Clinical and Experimental Medicine
| |
Collapse
|
36
|
Lim SH, Sun JM, Choi YL, Kim HR, Ahn S, Lee JY, Lee SH, Ahn JS, Park K, Kim JH, Cho BC, Ahn MJ. Efficacy and safety of dovitinib in pretreated patients with advanced squamous non-small cell lung cancer with FGFR1 amplification: A single-arm, phase 2 study. Cancer 2016; 122:3024-31. [PMID: 27315356 DOI: 10.1002/cncr.30135] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 11/08/2022]
Abstract
BACKGROUND Fibroblast growth factor receptor 1 (FGFR1) amplification is a potential driving oncogene in squamous cell cancer (SCC) of the lung. The current phase 2 study evaluated the efficacy and tolerability of dovitinib, an FGFR inhibitor, in patients with advanced SCC of the lung. METHODS Patients with pretreated advanced SCC of the lung whose tumors demonstrated FGFR1 amplification of > 5 copies by fluorescence in situ hybridization were enrolled. Dovitinib at a dose of 500 mg was administered orally, once daily, on days 1 to 5 of every week, followed by 2 days off. The primary endpoint was overall response. Secondary endpoints were progression-free survival, overall survival, and toxicity. RESULTS All 26 patients were men with a median age of 68 years (range, 52-80 years). The majority of patients were ever-smokers. The median duration of dovitinib administration (28 days per cycle) was 2.5 months (range, 0.7-8.6 months). The overall response rate was 11.5% (95% confidence interval [95% CI], 0.8%-23.8%) and the disease control rate was 50% (95% CI, 30.8%-69.2%), with 3 patients achieving partial responses. Response durations for the patients with partial responses were ≥4.5 months, ≥ 5.1 months, and 6.1 months, respectively. After a median follow-up of 15.7 months (range, 1.2-25.6 months), the median overall survival was 5.0 months (95% CI, 3.6-6.4 months) and the median progression-free survival was 2.9 months (95% CI, 1.5-4.3 months). The most common grade 3 or higher adverse events were fatigue (19.2%), anorexia (11.5%), and hyponatremia (11.5%) (event severity was graded based on National Cancer Institute Common Terminology Criteria for Adverse Events [version 4.0]). CONCLUSIONS Treatment with dovitinib demonstrated modest efficacy in patients with advanced SCC with FGFR1 amplification. Further studies to evaluate other biomarkers correlated with the efficacy of dovitinib in patients with SCC are warranted. Cancer 2016;122:3024-3031. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Sung Hee Lim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jong-Mu Sun
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hye Ryun Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Soomin Ahn
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Ji Yun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Keunchil Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Joo Hang Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Byoung Chul Cho
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea.
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.
| |
Collapse
|
37
|
Herrick CJ, Moley JF. New systemic therapies for locally advanced and metastatic thyroid cancer. INTERNATIONAL JOURNAL OF ENDOCRINE ONCOLOGY 2016. [DOI: 10.2217/ije-2015-0002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Thyroid cancer affects one in 100 people over their lifetime. Differentiated and medullary thyroid cancer, refractory to traditional therapy, respond poorly to chemotherapeutic agents. However, tyrosine kinase inhibitors provide new hope for stabilizing disease in patients with advanced progressive disease. There are multiple tyrosine kinase inhibitors under study for thyroid cancer and currently four drugs that are US FDA approved. Nonetheless, use of these drugs should be selective given a significant adverse event profile and diseases with a typically indolent course. This review will cover molecular mechanisms in thyroid cancer as they are relevant to targeted therapies and review available evidence for the safety and efficacy of therapies currently approved and under study for thyroid cancer.
Collapse
Affiliation(s)
- Cynthia J Herrick
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeffrey F Moley
- Section of Endocrine & Oncologic Surgery, Department of Surgery, Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
- Department of Surgery, St. Louis Veterans Affairs Medical Center, St. Louis, MO, USA
| |
Collapse
|