1
|
Wang Q, Pan G, Zhang Y, Ni Y, Mu Y, Luo D. Emerging insights into thyroid cancer from immunotherapy perspective: A bibliometric analysis. Hum Vaccin Immunother 2024; 20:2403170. [PMID: 39294892 DOI: 10.1080/21645515.2024.2403170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/21/2024] [Accepted: 09/07/2024] [Indexed: 09/21/2024] Open
Abstract
Thyroid cancer is a common endocrine malignancy that poses considerable therapeutic challenges in treating anaplastic carcinoma and advanced aggressive disease. Immunotherapy has become a prominent strategy for cancer treatment, and has shown remarkable advancements in recent years. In this study, we utilized visualization and bibliometric tools to analyze publications on thyroid cancer immunotherapy from the Web of Science Core Collection (WoSCC). A total of 409 articles were included, with an annual increase in both publications and citations since 2016. China leads research efforts in this area, while the University of Texas System and UTMD Anderson Cancer Center rank first in publication output. The journal Thyroid has garnered the highest citations. Notable authors contributing to this field include Antonelli Alessandro, Fallahi Poupak, and Wang Yu. Current research hotspots include immune checkpoint inhibitors, combination therapies involving immunotherapy with targeted therapy, CAR-T cell therapy, and modulation of the tumor microenvironment, all of which underscore the evolving landscape and potential for innovative treatments in thyroid cancer.
Collapse
Affiliation(s)
- Qianyu Wang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou, Zhejiang, China
| | - Gang Pan
- Department of Oncological Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Yu Zhang
- Department of Oncological Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Yiqin Ni
- Department of Oncological Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
| | - Yuzhu Mu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou, Zhejiang, China
| | - Dingcun Luo
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou First People's Hospital, Hangzhou, Zhejiang, China
- Department of Oncological Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, China
- College of Mathematical Medicine, Zhejiang Normal University, Jinhua, Zhejiang, China
| |
Collapse
|
2
|
Jannin A, Lugat A, Escande A, Godbert Y, Wasserman J, Cao CD, Hadoux J. Quelles avancées dans la prise en charge du carcinome anaplasique de la thyroïde en 2024 ? Bull Cancer 2024; 111:10S42-10S52. [PMID: 39505435 DOI: 10.1016/s0007-4551(24)00407-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2024]
Abstract
LATEST DEVELOPMENTS IN ANAPLASTIC THYROID CARCINOMA MANAGEMENT IN 2024: Anaplastic thyroid carcinomas (ATCs) represent a rare and undifferentiated form of thyroid cancer with a poor prognosis, typically marked by a median overall survival of four to ten months. However, recent advances have shown improvements due to the more systematic application of molecular testing, targeted therapies, and immunotherapy, alongside the establishment of rapid specialized care protocols in expert centers. Clinically, ATCs often present as a rapidly enlarging cervical mass originating from the thyroid, causing neck, pain and tenderness, dyspnea and dys-phagia, and associated lymphadenopathy, typically in elderly patients. Diagnostic confirmation requires an urgent biopsy, reviewed by a pathologist within the TUTHYREF-path network, with a mandatory search for BRAFV600E molecular alterations. Following diagnosis, care coordination is expedited within an expert center of the ENDOCAN-TUTHYREF network whenever feasible. Initial surgery is rarely an option due to frequent loco-regional cervical invasion. A multimodal treatment approach is essential: BRAF/MEK inhibitors (dabrafenib/trametinib) in cases of BRAFV600E mutation, or chemotherapy in the absence of a molecular target, combined with radiotherapy and, potentially, surgery if the disease becomes resectable following induction therapy. PD-1/PD-L1 targeted immunotherapy, either alone or in combination with targeted therapies, has shown potential to extend survival in some patients; however, predictive biomarkers and the optimal sequencing of immunotherapy (whether as induction and/or maintenance) require further clarification and may vary depending on the clinical context.
Collapse
Affiliation(s)
- Arnaud Jannin
- Université de Lille, CNRS, Inserm, CHU de Lille, UMR9020-U1277 - CANTHER - Hétérogénéité, Plasticité et Résistance aux Thérapies des Cancers, F-59000 Lille, France; CHU de Lille, Département d'endocrinologie, de diabétologie et de métabolisme, F-59000, Lille, France.
| | - Alexandre Lugat
- Département d'oncologie médicale, CHU de Nantes, 44000 Nantes, France; Département de médecine nucléaire, CHU de Nantes, 44000 Nantes, France; Centre de recherche sur le cancer CRCI2NA, Université de Nantes, Inserm UMR1307, CNRS-ERL6075, 44000 Nantes, France
| | - Alexandre Escande
- Service de radiothérapie, Centre Leonard-de-Vinci, 59187 Deuchy, France.
| | - Yann Godbert
- Institut Bergonié, Nuclear Medicine Department, 30003 Bordeaux, France
| | - Johanna Wasserman
- Service d'oncologie médicale, Hôpital universitaire Pitié-Salpêtrière, Assistance publique-Hôpitaux de Paris (AP-HP) Sorbonne université, 75013 Paris, France
| | - Christine Do Cao
- CHU de Lille, Département d'endocrinologie, de diabétologie et de métabolisme, F-59000, Lille, France
| | - Julien Hadoux
- Département d'imagerie, Service d'oncologie endocrinienne, Gustave-Roussy, Villejuif, F-94805, France.
| |
Collapse
|
3
|
Lin S, Cai H, Song X. Synergy between isobavachalcone and doxorubicin suppressed the progression of anaplastic thyroid cancer through ferroptosis activation. Braz J Med Biol Res 2024; 57:e13679. [PMID: 39166605 PMCID: PMC11338552 DOI: 10.1590/1414-431x2024e13679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/21/2024] [Indexed: 08/23/2024] Open
Abstract
The objective of this study was to explore the effects and mechanisms of the combination of isobavachalcone (IBC) and doxorubicin (DOX) on the progression of anaplastic thyroid cancer (ATC). Cell viability of 8505C and CAL62 cells was observed by CCK-8 assay. Kits were used to detect the presence of reactive oxygen species (ROS), glutathione (GSH), malondialdehyde (MDA), and cellular iron. Protein expression of solute carrier family 7 member 11 (SLC7A11) and glutathione peroxidase 4 (GPX4) was detected using western blot, and CD31 was detected through immunofluorescence. Tumor xenograft models of 8505C cells were constructed to observe the effect of IBC and DOX on ATC growth in vivo. The co-administration of IBC and DOX exhibited a synergistic effect of suppressing the growth of 8505C and CAL62 cells. The concurrent use of IBC and DOX resulted in elevated iron, ROS, and MDA levels, while reducing GSH levels and protein expression of SLC7A11 and GPX4. However, the Fer-1 ferroptosis inhibitor effectively counteracted this effect. In vitro and in vivo, the inhibitory effect on ATC cell proliferation and tumor growth was significantly enhanced by the combination of IBC and DOX. The combination of IBC and DOX can inhibit the growth of ATC by activating ferroptosis, and might prove to be a potent chemotherapy protocol for addressing ATC.
Collapse
Affiliation(s)
- Shuai Lin
- Department of Thyroid Breast Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Hui Cai
- Department of Thyroid Breast Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Xuemei Song
- Department of Operating Room, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| |
Collapse
|
4
|
Turner N, Hamidi S, Ouni R, Rico R, Henderson YC, Puche M, Alekseev S, Colunga-Minutti JG, Zafereo ME, Lai SY, Kim ST, Cabanillas ME, Nurieva R. Emerging therapeutic options for follicular-derived thyroid cancer in the era of immunotherapy. Front Immunol 2024; 15:1369780. [PMID: 38868771 PMCID: PMC11167082 DOI: 10.3389/fimmu.2024.1369780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/12/2024] [Indexed: 06/14/2024] Open
Abstract
Although most follicular-derived thyroid cancers are well differentiated and have an overall excellent prognosis following treatment with surgery and radioiodine, management of advanced thyroid cancers, including iodine refractory disease and poorly differentiated/undifferentiated subtypes, is more challenging. Over the past decade, better understanding of the genetic drivers and immune milieu of advanced thyroid cancers has led to significant progress in the management of these patients. Numerous targeted kinase inhibitors are now approved by the U.S Food and Drug administration (FDA) for the treatment of advanced, radioiodine refractory differentiated thyroid cancers (DTC) as well as anaplastic thyroid cancer (ATC). Immunotherapy has also been thoroughly studied and has shown promise in selected cases. In this review, we summarize the progress in the understanding of the genetic landscape and the cellular and molecular basis of radioiodine refractory-DTC and ATC, as well as discuss the current treatment options and future therapeutic avenues.
Collapse
Affiliation(s)
- Naimah Turner
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sarah Hamidi
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rim Ouni
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rene Rico
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ying C. Henderson
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Maria Puche
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Biology, College of Science and Engineering, Houston Christian University, Houston, TX, United States
| | - Sayan Alekseev
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Program of Biology, College of Sciences, The University of Texas at San Antonio, San Antonio, TX, United States
| | - Jocelynn G. Colunga-Minutti
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Program of Immunology, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX, United States
| | - Mark E. Zafereo
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Stephen Y. Lai
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Sang T. Kim
- Department of Rheumatology, Allergy and Immunology, Yale University, New Haven, CT, United States
| | - Maria E. Cabanillas
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Roza Nurieva
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Program of Immunology, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX, United States
| |
Collapse
|
5
|
Jungels C, Pita JM, Costante G. Anaplastic thyroid carcinoma: advances in molecular profiling and targeted therapy. Curr Opin Oncol 2023; 35:1-9. [PMID: 36398690 DOI: 10.1097/cco.0000000000000918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
PURPOSE OF REVIEW Anaplastic thyroid carcinomas (ATCs) are rare cancers with a globally very poor prognosis, because of their immensely aggressive behaviour, resulting in predominantly advanced stage of disease at diagnosis. Response to available therapies is still disappointing. Aim of the present review is to illustrate the diverse new strategies under investigation, to improve the poor outcome of these patients. RECENT FINDINGS Applying molecular analysis in ATC is unravelling potentially actionable targets of therapy. If a mutation of BRAF V600E is found, a combination of Dabrafenib and Trametinib is the recommended treatment. In the presence of another druggable mutation, a specific targeted therapy may be proposed. In the absence of druggable mutations, immunotherapy is an alternative approach, especially in case of significant PD-L1 expression. SUMMARY The molecular profiling of tumour samples is elucidating the genetic alterations involved in ATC development, and new preclinical models are under study to define innovative approaches for individualized treatment of such patients. Hopefully this approach could improve ATC prognosis.
Collapse
Affiliation(s)
- Christiane Jungels
- Department of Oncologic Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Bruxelles, Belgium
| | - Jaime Miguel Pita
- Institute of Interdisciplinary Research (IRIBHM) and ULB-Cancer Research Center (U-CRC), Université Libre de Bruxelles, Bruxelles, Belgium
| | - Giuseppe Costante
- Department of Oncologic Medicine, Institut Jules Bordet, Université Libre de Bruxelles, Bruxelles, Belgium
- Department of Endocrinology, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
6
|
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
|
7
|
Yadav D, Kwak M, Chauhan PS, Puranik N, Lee PCW, Jin JO. Cancer immunotherapy by immune checkpoint blockade and its advanced application using bio-nanomaterials. Semin Cancer Biol 2022; 86:909-922. [PMID: 35181474 DOI: 10.1016/j.semcancer.2022.02.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/09/2022] [Accepted: 02/13/2022] [Indexed: 02/07/2023]
Abstract
Cancer is the second leading cause of death worldwide. Traditional approaches, such as surgery, chemotherapy, and radiotherapy have been the main cancer therapeutic modalities in recent years. Cancer immunotherapy is a novel therapeutic modality that potentiates the immune responses of patients against malignancy. Immune checkpoint proteins expressed on T cells or tumor cells serve as a target for inhibiting T cell overactivation, maintaining the balance between self-reactivity and autoimmunity. Tumors essentially hijack the immune checkpoint pathway in order to survive and spread. Immune checkpoint inhibitors (ICIs) are being developed as a result to reactivate the anti-tumor immune response. Recent advances in nanotechnology have contributed to the development of successful, safe, and efficient anticancer drug systems based on nanoparticles. Nanoparticle-based cancer immunotherapy overcomes numerous challenges and offers novel strategies for improving conventional immunotherapies. The fundamental and physiochemical properties of nanoparticles depend on various cancer therapeutic strategies, such as chemotherapeutics, nucleic acid-based treatments, photothermal therapy, and photodynamic agents. The review discusses the use of nanoparticles as carriers for delivering immune checkpoint inhibitors and their efficacy in cancer combination therapy.
Collapse
Affiliation(s)
- Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea
| | - Minseok Kwak
- Department of Chemistry and Industry 4.0 Convergence Bionics Engineering, Pukyong National University, Busan, South Korea
| | | | - Nidhi Puranik
- Biological Sciences Department, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India
| | - Peter C W Lee
- Department of Biomedical Sciences, University of Ulsan College of Medicine, ASAN Medical Center, Seoul, South Korea.
| | - Jun-O Jin
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 38541, South Korea; Research Institute of Cell Culture, Yeungnam University, Gyeongsan, 38541, South Korea.
| |
Collapse
|
8
|
Zhang J, Veeramachaneni N. Targeting interleukin-1β and inflammation in lung cancer. Biomark Res 2022; 10:5. [PMID: 35086565 PMCID: PMC8796434 DOI: 10.1186/s40364-021-00341-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 11/03/2021] [Indexed: 12/16/2022] Open
Abstract
Inflammation is a process that protects organs against various potentially harmful stimuli and enables repair. Dysregulated inflammation, however, damages tissues and leads to disease, including cancer. Cancer-related inflammation is characterized by cytokine production, leukocyte infiltration, angiogenesis, and tissue remodeling-all critical processes in modulating the tumor microenvironment (TME). The TME is known to play a key role in tumor progression, and targeting its immune component to achieve a better anti-tumor response is the basis of immunotherapy. Despite the critical role cytokines play in the TME and tumor progression, there is currently only one therapy approved by the FDA that directly involves cytokine signaling: human recombinant interleukin-2 protein, aldesleukin. The recent Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS) trial evaluated the use of anti-interleukin-1β therapy in atherosclerotic disease; however, it also revealed interleukin-1β (IL-1β) blockade with canakinumab led to a significantly lower incidence of lung cancer. This has opened a promising new avenue for lung cancer therapy, and strategies using anti-IL-1β therapy alone or in combination with chemotherapy and/or immune checkpoint blockade are currently being evaluated in several clinical trials.
Collapse
Affiliation(s)
- Jun Zhang
- Division of Medical Oncology, Department of Internal Medicine, Department of Cancer Biology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.
| | - Nirmal Veeramachaneni
- Department of Cardiovascular and Thoracic Surgery, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160 , USA
| |
Collapse
|
9
|
Cavalieri S, Filippini DM, Ottini A, Bergamini C, Resteghini C, Colombo E, Lombardo R, Nuzzolese I, Alfieri S, Licitra L, Locati LD. Immunotherapy in head and neck squamous cell carcinoma and rare head and neck malignancies. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2021; 2:522-542. [PMID: 36046116 PMCID: PMC9400733 DOI: 10.37349/etat.2021.00062] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 10/29/2021] [Indexed: 11/19/2022] Open
Abstract
The dismal prognosis of recurrent/metastatic (R/M) head and neck squamous cell carcinoma (HNSCC) prompted recent advances in the field of therapeutic approaches beyond cytotoxic cancer therapy. In recent years, the deeper and increasing knowledge on the genomic landscape and the upcoming new data on immunotherapy enacted by HNSCCs have led to successful therapeutic targeting of the immune system. Immune checkpoint inhibitors (ICIs) have changed state of the art in R/M patients and could have a potential role even in early disease. The purpose of this work is to summarize the role of immunotherapy for R/M HNSCC in clinical practice, with insights about future perspectives. Updated immunotherapy results in other R/M head and neck cancers such as thyroid, salivary glands, nasopharynx, sinonasal cancers, and nuclear protein in testis (NUT) are presented.
Collapse
Affiliation(s)
- Stefano Cavalieri
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, via Festa del Perdono 7, 20122 Milan, Italy
| | - Daria Maria Filippini
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
| | - Arianna Ottini
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
| | - Cristiana Bergamini
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
| | - Carlo Resteghini
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
| | - Elena Colombo
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
| | - Roberta Lombardo
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
| | - Imperia Nuzzolese
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
| | - Salvatore Alfieri
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
| | - Lisa Licitra
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, via Festa del Perdono 7, 20122 Milan, Italy
| | - Laura D. Locati
- Head and Neck Cancer Medical Department, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, via Venezian 1, 20133 Milan, Italy
| |
Collapse
|
10
|
Nakhjavani M, Shigdar S. Future of PD-1/PD-L1 axis modulation for the treatment of triple-negative breast cancer. Pharmacol Res 2021; 175:106019. [PMID: 34861397 DOI: 10.1016/j.phrs.2021.106019] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 02/07/2023]
Abstract
Triple-negative breast cancer (TNBC) has the worst prognosis among the subtypes of breast cancer, with no targeted therapy available. Immunotherapy targeting programmed cell death protein-1 (PD-1) and its ligand (PD-L1) has resulted in some promising outcomes in cancer patients. The common treatments are monoclonal antibodies (mAbs). Despite novel methodologies in developing mAbs, there are several drawbacks with these medications. Immunological reactions, expensive and time-consuming production and requiring refrigeration are some of the challenging characteristics of mAbs that are addressed with using aptamers. Aptamers are nucleotide-based structures with high selectivity and specificity for target. Their small size helps aptamers penetrate the tissue better. In this review, we have discussed the nature of PD-1/PD-L1 interaction and summarised the available mAbs and aptamers specific for these two targets. This review highlights the role of aptamers as a future pathway for PD-1/PD-L1 modulation.
Collapse
Affiliation(s)
- Maryam Nakhjavani
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia; Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC 3220, Australia.
| | - Sarah Shigdar
- School of Medicine, Deakin University, Geelong, VIC 3220, Australia; Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, VIC 3220, Australia
| |
Collapse
|
11
|
Piha-Paul SA, Geva R, Tan TJ, Lim DW, Hierro C, Doi T, Rahma O, Lesokhin A, Luke JJ, Otero J, Nardi L, Singh A, Xyrafas A, Chen X, Mataraza J, Bedard PL. First-in-human phase I/Ib open-label dose-escalation study of GWN323 (anti-GITR) as a single agent and in combination with spartalizumab (anti-PD-1) in patients with advanced solid tumors and lymphomas. J Immunother Cancer 2021; 9:jitc-2021-002863. [PMID: 34389618 PMCID: PMC8365809 DOI: 10.1136/jitc-2021-002863] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2021] [Indexed: 11/29/2022] Open
Abstract
Background GWN323 is an IgG1 monoclonal antibody (mAb) against the glucocorticoid-induced tumor necrosis factor receptor-related protein. This first-in-human, open-label phase I/Ib study aimed to investigate the safety and tolerability and to identify the recommended doses of GWN323 with/without spartalizumab, an anti-programmed cell death receptor-1 agent, for future studies. Pharmacokinetics, preliminary efficacy and efficacy biomarkers were also assessed. Methods Patients (aged ≥18 years) with advanced/metastatic solid tumors with Eastern Cooperative Oncology Group performance status of ≤2 were included. GWN323 (10–1500 mg) or GWN323+spartalizumab (GWN323 10–750 mg+spartalizumab 100–300 mg) were administered intravenously at various dose levels and schedules during the dose-escalation phase. Dose-limiting toxicities (DLTs) were assessed during the first 21 days in a single-agent arm and 42 days in a combination arm. Adverse events (AEs) were graded per National Cancer Institute-Common Toxicity Criteria for Adverse Events V.4.03 and efficacy was assessed using Response Evaluation Criteria in Solid Tumors V.1.1. Results Overall, 92 patients (single-agent, n=39; combination, n=53) were included. The maximum administered doses (MADs) in the single-agent and combination arms were GWN323 1500 mg every 3 weeks (q3w) and GWN323 750 mg+spartalizumab 300 mg q3w, respectively. No DLTs were observed with single-agent treatment. Three DLTs (6%, all grade ≥3) were noted with combination treatment: blood creatine phosphokinase increase, respiratory failure and small intestinal obstruction. Serious AEs were reported in 30.8% and 34.0%, and drug-related AEs were reported in 82.1% and 77.4% of patients with single-agent and combination treatments, respectively. Disease was stable in 7 patients and progressed in 26 patients with single-agent treatment. In combination arm patients, 1 had complete response (endometrial cancer); 3, partial response (rectal cancer, adenocarcinoma of colon and melanoma); 14, stable disease; and 27, disease progression. GWN323 exhibited a pharmacokinetic profile typical of mAbs with a dose-dependent increase in the pharmacokinetic exposure. Inconsistent decreases in regulatory T cells and increases in CD8+ T cells were observed in the combination arm. Gene expression analyses showed no significant effect of GWN323 on interferon-γ or natural killer-cell signatures. Conclusions GWN323, as a single agent and in combination, was well tolerated in patients with relapsed/refractory solid tumors. The MAD was 1500 mg q3w for single-agent and GWN323 750 mg+spartalizumab 300 mg q3w for combination treatments. Minimal single-agent activity and modest clinical benefit were observed with the spartalizumab combination. Trial registration number NCT02740270.
Collapse
Affiliation(s)
- Sarina A Piha-Paul
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ravit Geva
- Division of Oncology, Tel-Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Tira J Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore.,Department of Medicine, Division of Medical Oncology and Hematology, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| | - Darren Wt Lim
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore
| | - Cinta Hierro
- Medical Oncology Department, Vall D'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain.,Molecular Therapeutics Research Unit (UITM), Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Toshikiko Doi
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
| | - Osama Rahma
- Center for Cancer Therapeutic Innovation, Gastrointestinal Cancer Center, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Alexander Lesokhin
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA.,Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jason John Luke
- Department of Hematology/Oncology, University of Chicago, Chicago, Illinois, USA.,Department of Hematology/Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Javier Otero
- Translational Clinical Oncology, Novartis Institutes for BioMedical Research Inc, East Hanover, New Jersey, USA
| | - Lisa Nardi
- Translational Clinical Oncology, Novartis Institutes for BioMedical Research Inc, East Hanover, New Jersey, USA
| | - Angad Singh
- Oncology Data Science, Novartis Institutes for BioMedical Research Inc, Cambridge, Massachusetts, USA
| | - Alexandros Xyrafas
- Early Development Analytics-Statistics, Novartis Pharma AG, Basel, Switzerland
| | - Xinhui Chen
- Oncology Therapeutic Area-Pharmacokinetic Sciences, Novartis Institutes for BioMedical Research Inc, East Hanover, New Jersey, USA
| | - Jennifer Mataraza
- Oncology Translational Research, Novartis Institutes for BioMedical Research Inc, Cambridge, Massachusetts, USA
| | - Philippe L Bedard
- Department of Medicine, Division of Medical Oncology and Hematology, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
12
|
Zheng L, Li L, He Q, Wang M, Ma Y, Zhu J, Li Y, Fu X, Zhang Y. Response to immunotherapy in a patient with anaplastic thyroid cancer: A case report. Medicine (Baltimore) 2021; 100:e26138. [PMID: 34397868 PMCID: PMC8360478 DOI: 10.1097/md.0000000000026138] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/25/2021] [Accepted: 05/11/2021] [Indexed: 01/04/2023] Open
Abstract
RATIONALE Anaplastic thyroid carcinoma (ATC) is an aggressive malignancy that is almost always fatal and lacks effective systemic treatment options. Current treatments of ATC include surgery, radiation, and chemotherapy, used in combination when possible. In the aspect of immunotherapy, the biomarker of TMB-H and MSI-H may suggest that patients benefit from pembrolizumab. Programmed cell death-ligand 1 (PD-L1) is highly expressed in ATC but has not been written into the guidelines or approved by the FDA as a biomarker for thyroid cancer immunotherapy. PATIENT CONCERNS A 55-year-old woman was admitted to our hospital because of a slight right-sided neck enlargement in November 2019. DIAGNOSES The clinical diagnosis was ATC, pT3bN0M0, and stage IVB. INTERVENTIONS Oral administration of apatinib (250 mg 3 times daily) was initiated after surgery, but some unpleasant side effects emerged after 1 month of treatment. Next-generation sequencing revealed that the tumor harbored 2 mutations, HRAS p.Q61R and TP53 p.P278S, and PD-L1 staining was positive with a high expression. Thus, camrelizumab (programmed cell death protein 1 inhibitor) was combined with apatinib, and apatinib was changed to 250 mg once a day from March 2020. OUTCOMES No adverse reactions were observed after the treatment immunotherapy combined with antiangiogenic drugs. Currently, the survival time of patients is more than 11 months, and the quality of life is not affected. CONCLUSION This case suggests that immunotherapy in patients with ATC based upon PD-L1 evaluation provides a therapeutic option. Targeting programmed cell death protein 1/PD-L1 may provide a much-needed treatment option for patients with advanced ATC.
Collapse
Affiliation(s)
- Luming Zheng
- Department of General Surgery, 960th Hospital of the People's Liberation Army, Jinan, Shandong, P. R. China
| | - Ling Li
- Yinfeng Gene Technology Co Ltd, CORA, Jinan, Shandong, P. R. China
| | - Qingqing He
- Department of General Surgery, 960th Hospital of the People's Liberation Army, Jinan, Shandong, P. R. China
| | - Meng Wang
- Department of General Surgery, 960th Hospital of the People's Liberation Army, Jinan, Shandong, P. R. China
| | - Yunhan Ma
- Department of General Surgery, 960th Hospital of the People's Liberation Army, Jinan, Shandong, P. R. China
| | - Jian Zhu
- Department of General Surgery, 960th Hospital of the People's Liberation Army, Jinan, Shandong, P. R. China
| | - Yanchen Li
- Department of General Surgery, 960th Hospital of the People's Liberation Army, Jinan, Shandong, P. R. China
| | - Xiaokang Fu
- Department of General Surgery, 960th Hospital of the People's Liberation Army, Jinan, Shandong, P. R. China
| | - Yaxuan Zhang
- Yinfeng Gene Technology Co Ltd, CORA, Jinan, Shandong, P. R. China
| |
Collapse
|
13
|
Dierks C, Seufert J, Aumann K, Ruf J, Klein C, Kiefer S, Rassner M, Boerries M, Zielke A, la Rosee P, Meyer PT, Kroiss M, Weißenberger C, Schumacher T, Metzger P, Weiss H, Smaxwil C, Laubner K, Duyster J, von Bubnoff N, Miething C, Thomusch O. Combination of Lenvatinib and Pembrolizumab Is an Effective Treatment Option for Anaplastic and Poorly Differentiated Thyroid Carcinoma. Thyroid 2021; 31:1076-1085. [PMID: 33509020 PMCID: PMC8290324 DOI: 10.1089/thy.2020.0322] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background: Anaplastic thyroid carcinoma (ATC) and metastatic poorly differentiated thyroid carcinomas (PDTCs) are rare aggressive malignancies with poor overall survival (OS) despite extensive multimodal therapy. These tumors are highly proliferative, with frequently increased tumor mutational burden (TMB) compared with differentiated thyroid carcinomas, and elevated programmed death ligand 1 (PD-L1) levels. These tumor properties implicate responsiveness to antiangiogenic and antiproliferative multikinase inhibitors such as lenvatinib, and immune checkpoint inhibitors such as pembrolizumab. Patients and Methods: In a retrospective study, we analyzed six patients with metastatic ATC and two patients with PDTC, who received a combination therapy of lenvatinib and pembrolizumab. Lenvatinib was started at 14-24 mg daily and combined with pembrolizumab at a fixed dose of 200 mg every three weeks. Maximum treatment duration with this combination was 40 months, and 3 of 6 ATC patients are still on therapy. Patient tumors were characterized by whole-exome sequencing and PD-L1 expression levels (tumor proportion score [TPS] 1-90%). Results: Best overall response (BOR) within ATCs was 66% complete remissions (4/6 CR), 16% stable disease (1/6 SD), and 16% progressive disease (1/6 PD). BOR within PDTCs was partial remission (PR 2/2). The median progression-free survival was 17.75 months for all patients, and 16.5 months for ATCs, with treatment durations ranging from 1 to 40 months (1, 4, 11, 15, 19, 25, 27, and 40 months). Grade III/IV toxicities developed in 4 of 8 patients, requiring dose reduction/discontinuation of lenvatinib. The median OS was 18.5 months, with three ATC patients being still alive without relapse (40, 27, and 19 months) despite metastatic disease at the time of treatment initiation (UICC and stage IVC). All patients with long-term (>2 years) or complete responses (CRs) had either increased TMB or a PD-L1 TPS >50%. Conclusions: Our results implicate that the combination of lenvatinib and pembrolizumab might be safe and effective in patients with ATC/PDTC and can result in complete and long-term remissions. The combination treatment is now being systematically examined in a phase II clinical trial (Anaplastic Thyroid Carcinoma Lenvatinib Pembrolizumab [ATLEP]) in ATC/PDTC patients.
Collapse
Affiliation(s)
- Christine Dierks
- Department of Hematology and Oncology, KIM IV, Faculty of Medicine, University Halle-Wittenberg, Halle, Germany
- Department of Hematology and Oncology, University of Freiburg, Freiburg, Germany
| | - Jochen Seufert
- Division of Endocrinology and Diabetology, Department of Medicine II, University of Freiburg, Freiburg, Germany
| | - Konrad Aumann
- Institute of Pathology, University of Freiburg, Germany
| | - Juri Ruf
- Department of Nuclear Medicine, University of Freiburg, Germany
| | - Claudius Klein
- Department of Hematology and Oncology, University of Freiburg, Freiburg, Germany
- Institute of Pathology, University of Freiburg, Germany
- Department of Nuclear Medicine, University of Freiburg, Germany
| | - Selina Kiefer
- Institute of Pathology, University of Freiburg, Germany
| | - Michael Rassner
- Department of Hematology and Oncology, University of Freiburg, Freiburg, Germany
| | - Melanie Boerries
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- Comprehensive Cancer Center Freiburg (CCCF), University Medical Center, University of Freiburg, Freiburg, Germany
- Institute of Medical Bioinformatics and Systems Medicine and Institute of Molecular Medicine and Cell Research; Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas Zielke
- Outcomes Research Unit, Department of Endocrine Surgery, Endocrine Center Stuttgart, Diakonie Klinikum Stuttgart, Stuttgart, Germany
| | - Paul la Rosee
- Klinikum Villingen-Schwenningen, Hämatologie/Onkologie, Villingen-Schwenningen, Germany
| | - Philipp Tobias Meyer
- Department of Nuclear Medicine, University of Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Matthias Kroiss
- Division of Endocrinology/Diabetology, Department of Internal Medicine, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Cancer Center Mainfranken, University of Würzburg, Würzburg, Germany
| | | | | | - Patrick Metzger
- Institute of Medical Bioinformatics and Systems Medicine and Institute of Molecular Medicine and Cell Research; Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | | | - Constantin Smaxwil
- Outcomes Research Unit, Department of Endocrine Surgery, Endocrine Center Stuttgart, Diakonie Klinikum Stuttgart, Stuttgart, Germany
| | - Katharina Laubner
- Division of Endocrinology and Diabetology, Department of Medicine II, University of Freiburg, Freiburg, Germany
| | - Justus Duyster
- Department of Hematology and Oncology, University of Freiburg, Freiburg, Germany
| | - Nikolas von Bubnoff
- Department of Hematology and Oncology, University of Freiburg, Freiburg, Germany
- Department of Hematology/Oncology, University of Luebeck, Luebeck, Germany
| | - Cornelius Miething
- Department of Hematology and Oncology, University of Freiburg, Freiburg, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
| | - Oliver Thomusch
- Department of General and Visceral Surgery, University Hospital Freiburg, Freiburg, Germany
| |
Collapse
|
14
|
Maniakas A, Dadu R, Busaidy NL, Wang JR, Ferrarotto R, Lu C, Williams MD, Gunn GB, Hofmann MC, Cote G, Sperling J, Gross ND, Sturgis EM, Goepfert RP, Lai SY, Cabanillas ME, Zafereo M. Evaluation of Overall Survival in Patients With Anaplastic Thyroid Carcinoma, 2000-2019. JAMA Oncol 2021; 6:1397-1404. [PMID: 32761153 DOI: 10.1001/jamaoncol.2020.3362] [Citation(s) in RCA: 183] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Importance Anaplastic thyroid carcinoma (ATC) historically has a 4-month median overall survival (OS) from time of diagnosis, with disease-specific mortality approaching 100%. The association between recent major advancements in treatment and OS has yet to be evaluated. Objective To evaluate rates of OS in patients with ATC over the last 2 decades. Design, Setting, and Participants Retrospective cohort study in a single tertiary care institution. Patients with histopathological confirmation of ATC from January 2000 to October 2019 were included and divided into 3 groups according to date of presentation: 2000-2013, 2014-2016, and 2017-2019. Main Outcomes and Measures Overall survival compared among different treatment eras and differing therapies, including targeted therapy, immunotherapy, and surgery. Results Of 479 patients (246 men [51%]; median age, 65.0 [range, 21.1-92.6] years) with ATC evaluated, 52 (11%) were stage IVA, 172 (36%) stage IVB, and 255 (53%) stage IVC at presentation. The median OS of the entire cohort was 0.79 years (9.5 months), ranging from 0.01 to 16.63. The OS at 1 and 2 years was 35% (95% CI, 29%-42%) and 18% (95% CI, 13%-23%) in the 2000-2013 group (n = 227), 47% (95% CI, 36%-56%) and 25% (95% CI, 17%-34%) in the 2014-2016 group (n = 100), and 59% (95% CI, 49%-67%) and 42% (95% CI, 30%-53%) in the 2017-2019 group (n = 152), respectively (P < .001). The hazard ratio was 0.50 (95% CI, 0.38-0.67) for the 2017-2019 group compared with the 2000-2013 patients (P < .001). Factors associated with improved OS included targeted therapy (hazard ratio, 0.49; 95% CI, 0.39-0.63; P < .001), the addition of immunotherapy to targeted therapy (hazard ratio, 0.58; 95% CI, 0.36-0.94; P = .03), and surgery following neoadjuvant BRAF-directed therapy (hazard ratio, 0.29; 95% CI, 0.10-0.78; P = .02). Patients undergoing surgery following neoadjuvant BRAF-directed therapy (n = 20) had a 94% 1-year survival with a median follow-up of 1.21 years. Conclusion and Relevance In this large single-institution cohort study spanning nearly 20 years, changes in patient management appear to be associated with significant increase in survival. The era of untreatable ATC is progressively being replaced by molecular-based personalized therapies, with integration of multidisciplinary therapies including surgery and radiation therapy.
Collapse
Affiliation(s)
- Anastasios Maniakas
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston
| | - Ramona Dadu
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston
| | - Naifa L Busaidy
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston
| | - Jennifer R Wang
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston
| | - Renata Ferrarotto
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Charles Lu
- Department of Thoracic Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Michelle D Williams
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston
| | - G Brandon Gunn
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Marie-Claude Hofmann
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston
| | - Gilbert Cote
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston
| | - Jared Sperling
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston
| | - Neil D Gross
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston
| | - Erich M Sturgis
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston
| | - Ryan P Goepfert
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston
| | - Stephen Y Lai
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston.,Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston
| | - Maria E Cabanillas
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston
| | - Mark Zafereo
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston
| |
Collapse
|
15
|
Minami H, Doi T, Toyoda M, Imamura Y, Kiyota N, Mitsuma A, Shimokata T, Naito Y, Matsubara N, Tajima T, Tokushige K, Ishihara K, Cameron S, Ando Y. Phase I study of the antiprogrammed cell death-1 Ab spartalizumab (PDR001) in Japanese patients with advanced malignancies. Cancer Sci 2021; 112:725-733. [PMID: 33031626 PMCID: PMC7893979 DOI: 10.1111/cas.14678] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/22/2020] [Accepted: 09/30/2020] [Indexed: 01/01/2023] Open
Abstract
Spartalizumab is a humanized IgG4/κ mAb directed against human programmed cell death-1 (PD-1). In this phase I study, we investigated safety, pharmacokinetics, preliminary antitumor activity, and toxicity of spartalizumab in patients with advanced malignancies. Patients (n = 18) with a range of tumor types received spartalizumab i.v. at doses of 1, 3, and 10 mg/kg every 2 weeks until disease progression, unacceptable toxicity, or discontinuation at the discretion of the investigator or patient. Most patients (61%) had received five or more prior lines of therapy. No dose-limiting toxicities were reported and, hence, the maximum tolerated dose was 10 mg/kg or more. Pharmacokinetics in Japanese patients aligned with those reported in a global dose-escalation study. The safety profile was consistent with other approved anti-PD-1 mAbs; the most common drug-related adverse events were maculopapular rash (22%), followed by malaise and increased blood alkaline phosphatase (11% each). Partial responses were reported in two patients (11%), one with transitional cell carcinoma and the other with hepatocellular carcinoma. In conclusion, this study confirmed the safety of spartalizumab given at a dose of up to 10 mg/kg every 2 weeks in Japanese patients with cancers.
Collapse
Affiliation(s)
- Hironobu Minami
- Kobe University Graduate School of Medicine and HospitalKobeJapan
| | | | - Masanori Toyoda
- Kobe University Graduate School of Medicine and HospitalKobeJapan
| | | | - Naomi Kiyota
- Kobe University Graduate School of Medicine and HospitalKobeJapan
| | | | | | - Yoichi Naito
- National Cancer Center Hospital EastKashiwaJapan
| | | | | | | | | | - Scott Cameron
- Novartis Institutes for BioMedical ResearchCambridgeMAUSA
| | | |
Collapse
|
16
|
Immune Checkpoint Blockade in Cancer Immunotherapy: Mechanisms, Clinical Outcomes, and Safety Profiles of PD-1/PD-L1 Inhibitors. Arch Immunol Ther Exp (Warsz) 2020; 68:36. [DOI: 10.1007/s00005-020-00601-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 10/31/2020] [Indexed: 02/07/2023]
|
17
|
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
|
18
|
Gray KD, McCloskey JE, Vedvyas Y, Kalloo OR, Eshaky SE, Yang Y, Shevlin E, Zaman M, Ullmann TM, Liang H, Stefanova D, Christos PJ, Scognamiglio T, Tassler AB, Zarnegar R, Fahey TJ, Jin MM, Min IM. PD1 Blockade Enhances ICAM1-Directed CAR T Therapeutic Efficacy in Advanced Thyroid Cancer. Clin Cancer Res 2020; 26:6003-6016. [PMID: 32887724 DOI: 10.1158/1078-0432.ccr-20-1523] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 07/17/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE Advanced thyroid cancers, including poorly differentiated and anaplastic thyroid cancer (ATC), are lethal malignancies with limited treatment options. The majority of patients with ATC have responded poorly to programmed death 1 (PD1) blockade in early clinical trials. There is a need to explore new treatment options. EXPERIMENTAL DESIGN We examined the expression of PD-L1 (a ligand of PD1) and intercellular adhesion molecule 1 (ICAM1) in thyroid tumors and ATC cell lines, and investigated the PD1 expression level in peripheral T cells of patients with thyroid cancer. Next, we studied the tumor-targeting efficacy and T-cell dynamics of monotherapy and combination treatments of ICAM1-targeting chimeric antigen receptor (CAR) T cells and anti-PD1 antibody in a xenograft model of ATC. RESULTS Advanced thyroid cancers were associated with increased expression of both ICAM1 and PD-L1 in tumors, and elevated PD1 expression in CD8+ T cells of circulating blood. The expression of ICAM1 and PD-L1 in ATC lines was regulated by the IFNγ-JAK2 signaling pathway. ICAM1-targeted CAR T cells, produced from either healthy donor or patient T cells, in combination with PD1 blockade demonstrated an improved ability to eradicate ICAM1-expressing target tumor cells compared with CAR T treatment alone. PD1 blockade facilitated clearance of PD-L1 high tumor colonies and curtailed excessive CAR T expansion, resulting in rapid tumor clearance and prolonged survival in a mouse model. CONCLUSIONS Targeting two IFNγ-inducible, tumor-associated antigens-ICAM1 and PD-L1-in a complementary manner might be an effective treatment strategy to control advanced thyroid cancers in vivo.
Collapse
Affiliation(s)
- Katherine D Gray
- Department of Surgery, Weill Cornell Medicine, New York, New York
| | | | - Yogindra Vedvyas
- Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Olivia R Kalloo
- Department of Surgery, Weill Cornell Medicine, New York, New York
| | - Steve El Eshaky
- Department of Surgery, Weill Cornell Medicine, New York, New York
| | - Yanping Yang
- Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Enda Shevlin
- Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Marjan Zaman
- Department of Radiology, Weill Cornell Medicine, New York, New York
| | | | - Heng Liang
- Department of Surgery, Weill Cornell Medicine, New York, New York
| | | | - Paul J Christos
- Department of Population Health Sciences, Weill Cornell Medicine, New York, New York
| | | | - Andrew B Tassler
- Department of Head and Neck Surgery, Weill Cornell Medicine, New York, New York
| | - Rasa Zarnegar
- Department of Surgery, Weill Cornell Medicine, New York, New York
| | - Thomas J Fahey
- Department of Surgery, Weill Cornell Medicine, New York, New York.
| | - Moonsoo M Jin
- Department of Radiology, Weill Cornell Medicine, New York, New York.
| | - Irene M Min
- Department of Surgery, Weill Cornell Medicine, New York, New York.
| |
Collapse
|
19
|
Abstract
OPINION STATEMENT Anaplastic thyroid cancer (ATC) is a rare but very aggressive form of undifferentiated thyroid cancer. Due to its rapid rate of progression and invasive nature, ATC poses significant risks of morbidity and mortality. The cornerstone in the management of ATC remains a prompt diagnosis of the disease and timely management of complications depending on the stage of disease. Surgery continues to offer a higher chance of a cure, although not all patients are candidates for surgical management. Patients with advanced disease may be considered for palliative surgery to reduce morbidity and complications from advanced disease. With the advent of new molecular testing and improved methods of diagnosis, novel therapeutic targets have been identified. Systemic therapy (chemotherapy and radiation therapy) as well as novel immunotherapy have shown some promise in patients with targetable genetic mutations. Patients should therefore have molecular testing of their tumor-if it is unresectable-and be tested for mutations that are targetable. Mutation-targeted therapy may be effective and may result in a significant response to allow surgical intervention for exceptional responders. Overall, patients who receive all three modalities of therapy (surgery, chemotherapy, and radiation therapy) have the highest overall survival.
Collapse
|
20
|
Yang J, Barletta JA. Anaplastic thyroid carcinoma. Semin Diagn Pathol 2020; 37:248-256. [PMID: 32624319 DOI: 10.1053/j.semdp.2020.06.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 06/22/2020] [Accepted: 06/22/2020] [Indexed: 02/06/2023]
Abstract
Anaplastic thyroid carcinoma (ATC) is a rare but significant malignancy due to its high mortality rate. Rendering an accurate diagnosis is crucial given the prognostic implications and treatment ramifications. Based on the prognostic significance of the extent of invasion of the primary tumor, T staging for ATC changed in the most recent edition of the American Joint Committee on Cancer (AJCC) staging manual. In the past 5 years there has been a rapid increase in our understanding of the molecular basis of ATC which has provided the basis for targeted therapy for some ATC patients. In this review, ATC prognostic factors, histologic and immunotypic features, staging updates, and molecular alterations, with an emphasis on those that may impact treatment, will be discussed.
Collapse
Affiliation(s)
- Jing Yang
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| | - Justine A Barletta
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| |
Collapse
|
21
|
Sukari A, Kukreja G, Nagasaka M, Shukairy MK, Yoo G, Lin HS, Hotaling J, Kim H. The role of immune checkpoint inhibitors in anaplastic thyroid cancer (Case Series). Oral Oncol 2020; 109:104744. [PMID: 32402656 DOI: 10.1016/j.oraloncology.2020.104744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 04/23/2020] [Indexed: 11/18/2022]
Abstract
Anaplastic thyroid carcinoma (ATC) is a rare type of thyroid neoplasm. However, it is one of the most aggressive forms of malignancy accounting for approximately 50% of mortality associated with all thyroid cancers. Here we report two cases of ATC treated with immune checkpoint inhibitors. Next generation sequencing identified BRAFV600E mutation in one of the patients who also derived benefit from BRAF targeted therapy. We here discuss these cases highlighting the importance of expert pathological review, utilizing molecular testing to identify the underlying genetic targets for personalized therapy, and the potential role of PD-1 inhibitors for the treatment of ATC.
Collapse
Affiliation(s)
- Ammar Sukari
- Department of Oncology, Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA.
| | - Geetika Kukreja
- Division of Hematology and Oncology, Department of Internal Medicine, Henry Ford Medical Group, Detroit, MI, USA
| | - Misako Nagasaka
- Department of Oncology, Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA; Division of Neurology, Department of Internal Medicine, St. Marianna University Graduate School of Medicine, Kawasaki, Japan
| | | | - George Yoo
- Department of Otolaryngology Head and Neck Surgery, Wayne State University, Detroit, MI, USA
| | - Ho-Sheng Lin
- Department of Otolaryngology Head and Neck Surgery, Wayne State University, Detroit, MI, USA
| | - Jeffrey Hotaling
- Department of Otolaryngology Head and Neck Surgery, Wayne State University, Detroit, MI, USA
| | - Harold Kim
- Division of Radiation Oncology, Department of Oncology, Karmanos Cancer Institute/Wayne State University, Detroit, MI, USA
| |
Collapse
|
22
|
Naing A, Gainor JF, Gelderblom H, Forde PM, Butler MO, Lin CC, Sharma S, Ochoa de Olza M, Varga A, Taylor M, Schellens JHM, Wu H, Sun H, Silva AP, Faris J, Mataraza J, Cameron S, Bauer TM. A first-in-human phase 1 dose escalation study of spartalizumab (PDR001), an anti-PD-1 antibody, in patients with advanced solid tumors. J Immunother Cancer 2020; 8:e000530. [PMID: 32179633 PMCID: PMC7073791 DOI: 10.1136/jitc-2020-000530] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2020] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Spartalizumab is a humanized IgG4κ monoclonal antibody that binds programmed death-1 (PD-1) and blocks its interaction with PD-L1 and PD-L2. This phase 1/2 study was designed to assess the safety, pharmacokinetics, and preliminary efficacy of spartalizumab in patients with advanced or metastatic solid tumors. METHODS In the phase 1 part of the study, 58 patients received spartalizumab, intravenously, at doses of 1, 3, or 10 mg/kg, administered every 2 weeks (Q2W), or 3 or 5 mg/kg every 4 weeks (Q4W). RESULTS Patients had a wide range of tumor types, most commonly sarcoma (28%) and metastatic renal cell carcinoma (10%); other tumor types were reported in ≤3 patients each. Most patients (93%) had received prior antineoplastic therapy (median three prior lines) and two-thirds of the population had tumor biopsies negative for PD-L1 expression at baseline. The maximum tolerated dose was not reached. The recommended phase 2 doses were selected as 400 mg Q4W or 300 mg Q3W. No dose-limiting toxicities were observed, and adverse events included those typical of other PD-1 antibodies. The most common treatment-related adverse events of any grade were fatigue (22%), diarrhea (17%), pruritus (14%), hypothyroidism (10%), and nausea (10%). Partial responses occurred in two patients (response rate 3.4%); one with atypical carcinoid tumor of the lung and one with anal cancer. Paired tumor biopsies from patients taken at baseline and on treatment suggested an on-treatment increase in CD8+ lymphocyte infiltration in patients with clinical benefit. CONCLUSIONS Spartalizumab was well tolerated at all doses tested in patients with previously treated advanced solid tumors. On-treatment immune activation was seen in tumor biopsies; however, limited clinical activity was reported in this heavily pretreated, heterogeneous population. The phase 2 part of this study is ongoing in select tumor types. TRIAL REGISTRATION NUMBER NCT02404441.
Collapse
Affiliation(s)
- Aung Naing
- MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | - Marcus O Butler
- Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| | - Chia-Chi Lin
- National Taiwan University Hospital, Taipei, Taiwan
| | - Sunil Sharma
- Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | | | | | - Matthew Taylor
- Oregon Health & Science University, Portland, Oregon, USA
| | | | - Hongqian Wu
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | - Haiying Sun
- Novartis Pharmaceuticals Corporation, East Hanover, New Jersey, USA
| | - Antonio P Silva
- Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Jason Faris
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | - Jennifer Mataraza
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | - Scott Cameron
- Novartis Institutes for BioMedical Research, Cambridge, Massachusetts, USA
| | - Todd M Bauer
- Sarah Cannon Research Institute, Nashville, Tennessee, USA
- Tennessee Oncology, PLLC, Nashville, Tennessee, USA
| |
Collapse
|
23
|
SEOM clinical guideline thyroid cancer (2019). Clin Transl Oncol 2020; 22:223-235. [PMID: 32006340 DOI: 10.1007/s12094-019-02284-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 12/27/2019] [Indexed: 12/13/2022]
Abstract
Thyroid carcinoma is the most frequent endocrine malignancy and accounts for around 3% of global cancer incidence. Different histologies and clinical scenarios make necessary a multidisciplinary approach that includes new diagnostic methods and surgical, radiopharmaceutical and systemic therapies. This guideline updates several aspects of management of thyroid cancer.
Collapse
|
24
|
Filetti S, Durante C, Hartl D, Leboulleux S, Locati LD, Newbold K, Papotti MG, Berruti A. Thyroid cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up†. Ann Oncol 2019; 30:1856-1883. [PMID: 31549998 DOI: 10.1093/annonc/mdz400] [Citation(s) in RCA: 572] [Impact Index Per Article: 114.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
MESH Headings
- Humans
- Adenocarcinoma, Follicular/diagnosis
- Adenocarcinoma, Follicular/epidemiology
- Adenocarcinoma, Follicular/pathology
- Adenocarcinoma, Follicular/therapy
- Carcinoma, Neuroendocrine/diagnosis
- Carcinoma, Neuroendocrine/epidemiology
- Carcinoma, Neuroendocrine/pathology
- Carcinoma, Neuroendocrine/therapy
- Follow-Up Studies
- Thyroid Cancer, Papillary/diagnosis
- Thyroid Cancer, Papillary/epidemiology
- Thyroid Cancer, Papillary/pathology
- Thyroid Cancer, Papillary/therapy
- Thyroid Carcinoma, Anaplastic/diagnosis
- Thyroid Carcinoma, Anaplastic/epidemiology
- Thyroid Carcinoma, Anaplastic/pathology
- Thyroid Carcinoma, Anaplastic/therapy
- Thyroid Neoplasms/diagnosis
- Thyroid Neoplasms/epidemiology
- Thyroid Neoplasms/pathology
- Thyroid Neoplasms/therapy
Collapse
Affiliation(s)
- S Filetti
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - C Durante
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - D Hartl
- Department of Head and Neck Oncology, Gustave Roussy, Villejuif; Université Paris Saclay, Villejuif
| | - S Leboulleux
- Université Paris Saclay, Villejuif; Department of Nuclear Medicine and Endocrine Oncology, Gustave Roussy, Villejuif, France
| | - L D Locati
- Head and Neck Medical Oncology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - K Newbold
- Head and Neck Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - M G Papotti
- Department of Pathology, University of Turin, Turin
| | - A Berruti
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Medical Oncology Unit, University of Brescia, ASST Spedali Civili, Brescia, Italy
| |
Collapse
|
25
|
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
|
26
|
Abstract
Although thyroid cancer generally has a good prognosis, there is a subset of patients for whom standard care (ie, treatment limited to surgery or surgery plus radioactive iodine) is either not appropriate because of the aggressive nature of their disease or not sufficient because of disease progression through standard treatment. Most of these tumors are in 3 groups: radioactive iodine-refractory differentiated thyroid carcinoma including poorly differentiated thyroid carcinoma anaplastic thyroid carcinoma, and progressive medullary thyroid carcinoma. Major classes of treatments in clinical development for these aggressive thyroid tumors include tyrosine kinase inhibitors, mammalian target of rapamycin inhibitors, and mitogen-activated protein kinase kinase inhibitors.
Collapse
Affiliation(s)
- Julian Huang
- Yale University School of Medicine, 123 York Street, 15A, New Haven, CT 06511, USA
| | - Ethan James Harris
- University of Illinois College of Medicine, 901 South Ashland Avenue, 01-715, Chicago, IL 60602, USA
| | - Jochen H Lorch
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, D2136, Boston, MA 02115, USA.
| |
Collapse
|
27
|
Dramatic Response to First Line Single Agent Pembrolizumab in Anaplastic Thyroid Carcinoma. Case Rep Endocrinol 2019; 2019:9095753. [PMID: 31885948 PMCID: PMC6899289 DOI: 10.1155/2019/9095753] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/08/2019] [Indexed: 11/20/2022] Open
Abstract
Anaplastic thyroid carcinoma (ATC) is a deadly disease with very limited therapeutic options. There is an urgent need for new and efficacious drugs. Unfortunately accrual in clinical trials is problematic because of the rarity of the disease and often poor performance status at diagnosis. Recently some data have emerged suggesting a role for immunotherapy in the treatment of ATC. We describe the case of a 75-year-old patient with poor performance status and compromised airway and oesophagus at diagnosis, showing a rapid and dramatic response to first line single agent pembrolizumab. Disease progression in the brain occurred 16 months after initial diagnosis. At that time there was ongoing extracranial disease control.
Collapse
|
28
|
Chintakuntlawar AV, Yin J, Foote RL, Kasperbauer JL, Rivera M, Asmus E, Garces NI, Janus JR, Liu M, Ma DJ, Moore EJ, Morris JC, Neben-Wittich M, Price DL, Price KA, Ryder M, Van Abel KM, Hilger C, Samb E, Bible KC. A Phase 2 Study of Pembrolizumab Combined with Chemoradiotherapy as Initial Treatment for Anaplastic Thyroid Cancer. Thyroid 2019; 29:1615-1622. [PMID: 31595822 DOI: 10.1089/thy.2019.0086] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Background: Anaplastic thyroid cancer (ATC) has poor prognosis with median overall survival (OS) of ∼6 months. We previously reported high PD-1/PDL-1 staining in ATC, raising the possibility of the productive application of the immunotherapeutic pembrolizumab. However, having found pembrolizumab to anecdotally have limited single-agent activity in ATC, we sought to alternatively define whether pembrolizumab might synergistically combine with chemoradiotherapy as initial ATC therapy. Methods: An investigator-initiated therapeutic phase 2 trial of pembrolizumab, 200 mg intravenously (IV) every 3 weeks, combined with chemoradiotherapy (docetaxel/doxorubicin, 20 mg/m2 each IV weekly plus volumetric modulated arc therapy) was initiated as frontline therapy (with or without surgery) in ATC to assess efficacy and toxicities. Six-month OS was selected as the primary endpoint using a Simon's optimal design with interim analysis (targeting accrual of 25 patients; Cohort A: prior resection, Cohort B: no resection). Based on a prior patient cohort-treated similarly, but without pembrolizumab, the design was such that, if 6-month true survival is 75%, the probability of declaring the approach worthy of further pursuit would be 91%. Results: Three patients were enrolled, two with rapidly enlarging unresectable neck masses. Early tumor responses were favorable in all three, and all three satisfactorily completed: intended radiotherapy, preceding and radiotherapy-concurrent pembrolizumab, and concurrent chemoradiotherapy. However, all three patients died <6 months following therapy initiation-one from pulmonary metastases and two from otherwise unexpected fatal pulmonary complications occurring subsequent to chemoradiotherapy completion-prompting study closure. Conclusions: Although initially tolerated and effective in terms of locoregional disease control, disappointing survival outcomes compared with historical controls raise uncertainty that the piloted approach merits further pursuit in ATC.
Collapse
Affiliation(s)
| | - Jun Yin
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Robert L Foote
- Department of Radiation Oncology and Mayo Clinic, Rochester, Minnesota
| | - Jan L Kasperbauer
- Division of Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota
| | - Michael Rivera
- Department of Laboratory and Anatomical Pathology, Mayo Clinic, Rochester, Minnesota
| | - Erik Asmus
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Nina I Garces
- Department of Radiation Oncology and Mayo Clinic, Rochester, Minnesota
| | - Jeffrey R Janus
- Division of Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota
| | - Minetta Liu
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Daniel J Ma
- Department of Radiation Oncology and Mayo Clinic, Rochester, Minnesota
| | - Eric J Moore
- Division of Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota
| | - John C Morris
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
- Division of Endocrinology, Mayo Clinic, Rochester, Minnesota
| | | | - Daniel L Price
- Division of Head and Neck Surgery, Mayo Clinic, Rochester, Minnesota
| | | | - Mabel Ryder
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
- Division of Endocrinology, Mayo Clinic, Rochester, Minnesota
| | | | - Crystal Hilger
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Eleyna Samb
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| | - Keith C Bible
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
29
|
Pembrolizumab for anaplastic thyroid cancer: a case study. Cancer Immunol Immunother 2019; 68:1921-1934. [PMID: 31637475 DOI: 10.1007/s00262-019-02416-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 10/10/2019] [Indexed: 12/18/2022]
Abstract
Blockade of the PD-1/PD-L1 pathway with targeted monoclonal antibodies has demonstrated encouraging anti-tumour activity in multiple cancer types. We present the case of a patient with BRAF-negative stage IVC anaplastic thyroid cancer (ATC) treated with the anti-PD-1 monoclonal antibody, pembrolizumab, following radiographic progression on chemoradiation. Blood samples were collected prior to and at four time points during treatment with pembrolizumab. Mass cytometry was used to determine expression of relevant biomarkers by peripheral blood mononuclear cells. Faecal samples were collected at baseline and 4 weeks following treatment initiation; taxonomic profiling using 16S ribosomal RNA (rRNA) gene sequencing was performed. Following treatment, a marked expansion in CD20+ B cell, CD16+ CD56lo NK cell and CD45RO+ CCR7+ central memory CD4+ T-cell populations was observed in the peripheral blood. Proportions of cells expressing the co-receptors TIGIT, OX40 and CD86 also increased during treatment. A high abundance of bacteria of the order Bacteroidales, specifically from the Bacteroidaceae and Rikenellaceae families, was identified in the faecal microbiota. Moreover, the patient's microbiome was enriched in Clostridiales order members Ruminococcaceae, Veillonellaceae and Lachnospiraceae. Alpha diversity of the gut microbiome was significantly higher following initiation of checkpoint therapy as assessed by the Shannon and Simpson index. Our results suggest that treatment with pembrolizumab promotes expansion of T-, B- and NK cell populations in the peripheral blood at the time of tumour regression and have the potential to be implemented as predictive biomarkers in the context of checkpoint blockade therapy. Larger studies to confirm these findings are warranted.
Collapse
|
30
|
Anaplastic Thyroid Cancer: Clinical Picture of the Last Two Decades at a Single Oncology Referral Centre and Novel Therapeutic Options. Cancers (Basel) 2019; 11:cancers11081188. [PMID: 31443283 PMCID: PMC6721627 DOI: 10.3390/cancers11081188] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Revised: 08/05/2019] [Accepted: 08/09/2019] [Indexed: 12/31/2022] Open
Abstract
Anaplastic thyroid cancer (ATC) is a rare tumour but also one of the most lethal malignancies. Therapeutic modalities have usually been limited, but clinical trials with new drugs are now being implemented. The aims of this study were to analyse the clinical presentation, therapeutic modalities and independent prognostic factors for survival. We also reviewed the most recent literature on novel ATC therapies. We performed a retrospective analysis of 79 patients diagnosed between 2000 and 2018. Variables with impact on survival were identified using the Cox proportional-hazard regression model. At presentation, 6.3% had thyroid-confined disease, 30.4% evidenced extrathyroidal extension and 60.8% were already metastatic. Surgery was feasible in 41.8% and radiotherapy was applied to 35.4%, with those receiving >45 Gy having longer estimated survival (p = 0.020). Chemotherapy, either conventional or with tyrosine kinase inhibitors, was performed in 17.7% and 7.6%, respectively. Multimodality therapy with surgery, radiotherapy and chemotherapy/tyrosine kinase inhibitors (TKI) had the greatest impact on disease specific survival (DSS), providing a risk reduction of death of 96.9% (hazard ratio (HR) = 0.031, 0.005–0.210, p < 0.001). We concluded that most of these patients join reference centres at advanced stages of disease and multimodality treatment may offer the best chances for prolonging survival.
Collapse
|
31
|
Cohen EEW, Bell RB, Bifulco CB, Burtness B, Gillison ML, Harrington KJ, Le QT, Lee NY, Leidner R, Lewis RL, Licitra L, Mehanna H, Mell LK, Raben A, Sikora AG, Uppaluri R, Whitworth F, Zandberg DP, Ferris RL. The Society for Immunotherapy of Cancer consensus statement on immunotherapy for the treatment of squamous cell carcinoma of the head and neck (HNSCC). J Immunother Cancer 2019; 7:184. [PMID: 31307547 PMCID: PMC6632213 DOI: 10.1186/s40425-019-0662-5] [Citation(s) in RCA: 417] [Impact Index Per Article: 83.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 07/02/2019] [Indexed: 02/06/2023] Open
Abstract
Head and neck cancers, including those of the lip and oral cavity, nasal cavity, paranasal sinuses, oropharynx, larynx and nasopharynx represent nearly 700,000 new cases and 380,000 deaths worldwide per annum, and account for over 10,000 annual deaths in the United States alone. Improvement in outcomes are needed for patients with recurrent and or metastatic squamous cell carcinoma of the head and neck (HNSCC). In 2016, the US Food and Drug Administration (FDA) granted the first immunotherapeutic approvals - the anti-PD-1 immune checkpoint inhibitors nivolumab and pembrolizumab - for the treatment of patients with recurrent squamous cell carcinoma of the head and neck (HNSCC) that is refractory to platinum-based regimens. The European Commission followed in 2017 with approval of nivolumab for treatment of the same patient population, and shortly thereafter with approval of pembrolizumab monotherapy for the treatment of recurrent or metastatic HNSCC in adults whose tumors express PD-L1 with a ≥ 50% tumor proportion score and have progressed on or after platinum-containing chemotherapy. Then in 2019, the FDA granted approval for PD-1 inhibition as first-line treatment for patients with metastatic or unresectable, recurrent HNSCC, approving pembrolizumab in combination with platinum and fluorouracil for all patients with HNSCC and pembrolizumab as a single agent for patients with HNSCC whose tumors express a PD-L1 combined positive score ≥ 1. These approvals marked the first new therapies for these patients since 2006, as well as the first immunotherapeutic approvals in this disease. In light of the introduction of these novel therapies for the treatment of patients with head and neck cancer, The Society for Immunotherapy of Cancer (SITC) formed an expert committee tasked with generating consensus recommendations for emerging immunotherapies, including appropriate patient selection, therapy sequence, response monitoring, adverse event management, and biomarker testing. These consensus guidelines serve as a foundation to assist clinicians' understanding of the role of immunotherapies in this disease setting, and to standardize utilization across the field for patient benefit. Due to country-specific variances in approvals, availability and regulations regarding the discussed agents, this panel focused solely on FDA-approved drugs for the treatment of patients in the U.S.
Collapse
Affiliation(s)
- Ezra E W Cohen
- Moores Cancer Center, University of California San Diego, San Diego, CA, USA
| | - R Bryan Bell
- Earle A. Chiles Research Institute at the Robert W. Franz Cancer Center, Providence Cancer Institute, Portland, OR, USA
| | - Carlo B Bifulco
- Earle A. Chiles Research Institute at the Robert W. Franz Cancer Center, Providence Cancer Institute, Portland, OR, USA
| | - Barbara Burtness
- Yale School of Medicine and Yale Cancer Center, New Haven, CT, USA
| | - Maura L Gillison
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Nancy Y Lee
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rom Leidner
- Earle A. Chiles Research Institute at the Robert W. Franz Cancer Center, Providence Cancer Institute, Portland, OR, USA
| | | | - Lisa Licitra
- Fondazione IRCCS Istituto Nazionale dei Tumori Milan and University of Milan, Milan, Italy
| | - Hisham Mehanna
- Institute of Head and Neck Studies and Education, University of Birmingham, Birmingham, UK
| | - Loren K Mell
- Moores Cancer Center, University of California San Diego, San Diego, CA, USA
| | - Adam Raben
- Helen F. Graham Cancer Center, Newark, DE, USA
| | | | - Ravindra Uppaluri
- Brigham and Women's Hospital and Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | | |
Collapse
|
32
|
Chintakuntlawar AV, Foote RL, Kasperbauer JL, Bible KC. Diagnosis and Management of Anaplastic Thyroid Cancer. Endocrinol Metab Clin North Am 2019; 48:269-284. [PMID: 30717908 DOI: 10.1016/j.ecl.2018.10.010] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Anaplastic thyroid cancer (ATC) is a devastating and usually incurable diagnosis. Clinical and pathologic diagnosis is best assessed at a tertiary center with concentrated ATC expertise. Expeditious multidisciplinary management is recommended for optimal patient outcomes. Based on multiinstitutional and population-based studies, multimodal therapy that includes chemoradiotherapy with surgery (when feasible) is the preferred initial treatment because it is associated with incrementally improved overall survival. In ATC that carries a BRAF V600E somatic mutation, combination therapy with BRAF and MEK inhibitors has shown promise but needs further study. Immunotherapeutic agents in neoadjuvant and metastatic settings are being investigated.
Collapse
Affiliation(s)
| | - Robert L Foote
- Department of Radiation Oncology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Jan L Kasperbauer
- Division of Head and Neck Surgery, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA
| | - Keith C Bible
- Division of Medical Oncology, Mayo Clinic, 200 First Street Southwest, Rochester, MN 55905, USA.
| |
Collapse
|
33
|
Khan SA, Ci B, Xie Y, Gerber DE, Beg MS, Sherman SI, Cabanillas ME, Busaidy NL, Burtness BA, Heilmann AM, Bailey M, Ross JS, Sher DJ, Ali SM. Unique mutation patterns in anaplastic thyroid cancer identified by comprehensive genomic profiling. Head Neck 2019; 41:1928-1934. [PMID: 30758123 DOI: 10.1002/hed.25634] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 08/27/2018] [Accepted: 12/19/2018] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION Anaplastic thyroid cancer (ATC) is a highly aggressive thyroid cancer. Those ATC with genomic alterations (GAs) in TSC2, ALK, and BRAF may respond to targeted therapies. METHODS Comprehensive genomic profiling on 90 ATC specimens identified base substitutions, short insertions and deletions, amplifications, copy number alterations, and genomic rearrangements in up to 315 cancer-related genes and 28 genes commonly rearranged in cancer. RESULTS Median patient age was 65 (range, 33-86) years, 50 patients were male. There was a mean of 4.2 GA per case, range 1-11. The most common GA were TP53 (66%), BRAF (34%), TERT (32%), CDKN2A (32%), and NRAS (26%). BRAF V600E and NRAS/HRAS/KRAS alteration were mutually exclusive. BRAF, CDKN2A, PIK3CA, and JAK2 were more frequent in patients >70 years of age; while myc, PTEN, and NRAS were more common in those ≤50 years. CONCLUSION ATC shows many GA with potential therapeutic significance and suggesting different molecular pathways can lead to ATC.
Collapse
Affiliation(s)
- Saad A Khan
- Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Bo Ci
- Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yang Xie
- Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - David E Gerber
- Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Clinical Sciences, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Muhammad S Beg
- Division of Hematology and Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Steven I Sherman
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maria E Cabanillas
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naifa L Busaidy
- Department of Endocrine Neoplasia and Hormonal Disorders, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Barbara A Burtness
- Department of Internal Medicine, Yale School of Medicine; Developmental Therapeutics Program, Yale Cancer Center, New Haven, Connecticut
| | | | - Mark Bailey
- Foundation Medicine, Inc., Cambridge, Massachusetts
| | | | - David J Sher
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Siraj M Ali
- Foundation Medicine, Inc., Cambridge, Massachusetts
| |
Collapse
|
34
|
Möckelmann N, Münscher A. [New aspects in thyroid cancer treatment : Highlights of the ASCO Annual Meeting 2018]. HNO 2018; 66:896-900. [PMID: 30402812 DOI: 10.1007/s00106-018-0589-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Whereas surgical treatment of thyroid carcinoma plays an important role especially in the early stages, a multimodal approach is pursued in the palliative setting, which, in addition to classical chemotherapy primarily involves treatment with tyrosine kinase inhibitors. An analysis of clinical trials and studies presented at the American Society of Clinical Oncology (ASCO) Annual Meeting 2018 is presented. In particular, studies on the treatment of radioiodine-refractory differentiated thyroid cancer, anaplastic thyroid cancer and immunotherapy were selected and analyzed. Clinically and preclinically relevant studies are presented and critically interpreted in this review.
Collapse
Affiliation(s)
- N Möckelmann
- Klinik und Poliklinik für Hals‑, Nasen- und Ohrenheilkunde, Kopf-Hals Chirurgie, Kopf-Hals-Tumorzentrum des Universitären Cancer Center Hamburg (UCCH), Universitätsklinikum Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Deutschland.
| | - A Münscher
- Klinik und Poliklinik für Hals‑, Nasen- und Ohrenheilkunde, Kopf-Hals Chirurgie, Kopf-Hals-Tumorzentrum des Universitären Cancer Center Hamburg (UCCH), Universitätsklinikum Hamburg-Eppendorf, Martinistraße 52, 20246, Hamburg, Deutschland
| |
Collapse
|
35
|
Gunda V, Gigliotti B, Ndishabandi D, Ashry T, McCarthy M, Zhou Z, Amin S, Freeman GJ, Alessandrini A, Parangi S. Combinations of BRAF inhibitor and anti-PD-1/PD-L1 antibody improve survival and tumour immunity in an immunocompetent model of orthotopic murine anaplastic thyroid cancer. Br J Cancer 2018; 119:1223-1232. [PMID: 30327563 PMCID: PMC6251038 DOI: 10.1038/s41416-018-0296-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 08/17/2018] [Accepted: 09/19/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Patients with anaplastic thyroid cancer (ATC) have an extremely poor prognosis despite aggressive multimodal therapy. ATC has a high prevalence of BRAFV600E mutations and is associated with an immunosuppressive microenvironment; we previously demonstrated that the combination of BRAF inhibitor and checkpoint inhibitor immunotherapy synergistically reduce tumour volume in an immunocompetent mouse model of orthotopic ATC. METHODS We again utilised our mouse model of ATC to assess the combination of BRAFV600E inhibitor PLX4720 and anti-PD-L1 or anti-PD-1 antibody on survival, and performed immune cell profiling of lymphoid and myeloid-lineage cells during maximal treatment response and tumour regrowth. RESULTS Combination therapy dramatically improved mouse survival. Maximal tumour reduction was associated with increases in the number and cytotoxicity of CD8+ T cells and NK cells, as well as increases in mostly M1-polarised tumour-associated macrophages (TAM) and decreases in myeloid-derived suppressor-like cells. Regrowth of tumour occurred after 2-3 weeks of ongoing combination therapy, and was most significantly associated with decreased TAMs and a dramatic increase in M2-polarisation. CONCLUSIONS Combination of PLX4720 and anti-PD-L1/PD-1 antibody dramatically reduced tumour volume, prolonged survival and improved the anti-tumour immune profile in murine ATC. Tumour growth inevitably recurred and demonstrated re-emergence of an immunosuppressive tumour microenvironment.
Collapse
Affiliation(s)
- Viswanath Gunda
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Benjamin Gigliotti
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dorothy Ndishabandi
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tameem Ashry
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael McCarthy
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Zhiheng Zhou
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Salma Amin
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alessandro Alessandrini
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sareh Parangi
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
36
|
Rao SN, Cabanillas ME. Navigating Systemic Therapy in Advanced Thyroid Carcinoma: From Standard of Care to Personalized Therapy and Beyond. J Endocr Soc 2018; 2:1109-1130. [PMID: 30250937 PMCID: PMC6141902 DOI: 10.1210/js.2018-00180] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/08/2018] [Indexed: 12/13/2022] Open
Abstract
Thyroid cancer, with the exception of anaplastic thyroid cancer, typically has very favorable outcomes with the standard therapy. However, those that persist, recur, or metastasize are associated with a worse prognosis. Targeted therapy with kinase inhibitors has shown promise in advanced cases of thyroid cancer, and currently five drug regimens are approved for use in clinical practice in the treatment of differentiated, medullary, and anaplastic thyroid cancer, with more options in the pipeline. However, one of the greatest dilemmas is when and how to initiate one of these drugs, and this is discussed herein.
Collapse
Affiliation(s)
- Sarika N Rao
- MedStar Washington Hospital Center, Washington, DC
- Georgetown University School of Medicine, Washington, DC
| | | |
Collapse
|
37
|
Iyer PC, Dadu R, Gule-Monroe M, Busaidy NL, Ferrarotto R, Habra MA, Zafereo M, Williams MD, Gunn GB, Grosu H, Skinner HD, Sturgis EM, Gross N, Cabanillas ME. Salvage pembrolizumab added to kinase inhibitor therapy for the treatment of anaplastic thyroid carcinoma. J Immunother Cancer 2018; 6:68. [PMID: 29996921 PMCID: PMC6042271 DOI: 10.1186/s40425-018-0378-y] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 06/20/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Anaplastic thyroid carcinoma (ATC) is a rare but deadly form of thyroid cancer. Kinase inhibitors kinase inhibitors have shown clinical efficacy in the management of ATC, however, eventually these tumors acquire resistance to KI and patients succumb to their disease. Salvage therapy in this setting is limited. As ATC tumors diffusely express the programmed cell death protein ligand (PD-L1), anti- programmed cell death protein (PD-1) drugs such as pembrolizumab offer therapeutic potential. We sought to explore the efficacy of adding pembrolizumab to kinase inhibitors at progression in ATC. METHODS We retrospectively reviewed the charts of ATC patients initiated on pembrolizumab in combination with KI at the time of progression on kinase inhibitors at MD Anderson Cancer Center between August 2016 and August 2017. Efficacy was evaluated with best overall response (BOR) using RECISTv1.1 criteria. Progression free survival (PFS) from the start of pembrolizumab and overall survival (OS) from the start of kinase inhibitors, as well as from the time of addition of pembrolizumab were calculated. RESULTS Twelve patients were treated with combination kinase inhibitors plus pembrolizumab at the time of progression on their KI therapy. Median age at initiation of pembrolizumab was 60 years (range 47-84 years). BOR was as follows: 5/12 (42%) had partial response, 4/12 (33%) had stable disease and 3/12 (25%) had progressive disease. Median OS from the start of kinase inhibitor was 10.43 months (95% CI = 6.02, 14.83, range 5.4-40 months). Median OS and PFS from the addition of pembrolizumab were 6.93 months (95% CI = 1.7, 12.15, range 3-15.9 months) and 2.96 months (95% CI = 2.2, 3.7, range 0.57-13.14 months), respectively. Fatigue, anemia and hypertension were the most common AEs encountered on these combinations. Therapy had to be discontinued in 2 patients due to drug induced rash and altered mental status likely from progression of disease. CONCLUSION In a subset of ATC patients, pembrolizumab may be an effective salvage therapy added to kinase inhibitors at the time of progression on these drugs. However, better treatment strategies aimed at incorporating immunotherapy in patients with ATC should be explored. Frontline combination of KI with immunotherapy should be studied in prospective clinical trials.
Collapse
Affiliation(s)
- Priyanka C. Iyer
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1461, Houston, TX 77030 USA
| | - Ramona Dadu
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1461, Houston, TX 77030 USA
| | - Maria Gule-Monroe
- Department of Diagnostic Radiology, Division of Diagnostic Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1482, Houston, TX 77030 USA
| | - Naifa L. Busaidy
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1461, Houston, TX 77030 USA
| | - Renata Ferrarotto
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 0432, Houston, TX 77030 USA
| | - Mouhammed Amir Habra
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1461, Houston, TX 77030 USA
| | - Mark Zafereo
- Department of Head and Neck Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1445, Houston, TX 77030 USA
| | - Michelle D. Williams
- Department of Pathology, Division of Pathology/Lab Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 0085, Houston, TX 77030 USA
| | - G. Brandon Gunn
- Department of Radiation Oncology, Division of Radiation Oncology, Proton Therapy Center, 1515 Holcombe Blvd, Unit 0097, Houston, TX USA
| | - Horiana Grosu
- Department of Pulmonary Medicine, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1462, Houston, TX 77030 USA
| | - Heath D. Skinner
- Department of Radiation Oncology, Division of Radiation Oncology, Proton Therapy Center, 1515 Holcombe Blvd, Unit 0097, Houston, TX USA
| | - Erich M. Sturgis
- Department of Head and Neck Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1445, Houston, TX 77030 USA
| | - Neil Gross
- Department of Head and Neck Surgery, Division of Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1445, Houston, TX 77030 USA
| | - Maria E. Cabanillas
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1461, Houston, TX 77030 USA
| |
Collapse
|