1
|
Ashayeri N, Khani P, Miri-Aliabad G, Jafari M, Pajouhi A. Malignant melanoma with bone metastases in a child: a case report and review of literature. Ann Med Surg (Lond) 2025; 87:318-322. [PMID: 40109597 PMCID: PMC11918636 DOI: 10.1097/ms9.0000000000002767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/04/2024] [Indexed: 03/22/2025] Open
Abstract
Introduction and importance Melanoma is the fifth most common cancer diagnosed in the US in 2022. While its incidence increased from 1980 to 2017, it rarely happens in children. Its diagnosis and treatment are challenging in pediatric patients due to its rarity and different presentations compared to adults. Case presentation An 11.5-year-old boy with a complaint of back pain was admitted to the hospital. Initial examinations were normal; however, due to continued pain and symptoms (e.g., inability to walk, knee pain and ankle ecchymosis, weight loss, vomiting, nausea, and dyspnea), further examinations were performed. Biopsy, nuclear scan, computed tomography (CT) scan, and immunohistochemistry (IHC) confirmed the malignant melanoma diagnosis with bone and lung metastasis. Single-agent treatment with nivolumab was initiated after the diagnosis confirmation with no specific complication. The patient's parents decided to continue the treatment in their city of residence to reduce costs. However, the treatment was not continued, and unfortunately, the patient has passed away. Clinical discussion Melanoma is rare in children, making its diagnosis challenging. There are no specific guidelines for treating melanoma in pediatric patients, especially in children under 12, with ongoing debate on the most suitable treatment and follow-up options for these patients. It is important to fully examine resected legions for malignancy. Conclusion The study highlights the difficulties in diagnosing and treating malignant melanoma in children, considering its rarity and unusual signs and symptoms compared to adults. Specific guidelines are needed for diagnosing and treating malignant melanoma in pediatric patients.
Collapse
Affiliation(s)
- Neda Ashayeri
- Department of Pediatric Hematology and Oncology, Hazrat-e Ali Asghar Children's Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Parya Khani
- Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Ghasem Miri-Aliabad
- Department of Pediatric Hematology and Oncology, Hazrat-e Ali Asghar Children's Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Jafari
- Department of Pediatric Hematology and Oncology, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Ali Pajouhi
- Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran
- USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
2
|
Moscardini-Martelli J, Rodríguez-Camacho A, Torres-Ríos JA, Meraz-Soto JM, Flores-Vázquez JG, Hernández-Sánchez LC, Lozano-Ruiz FJ, Maldonado-Magos F, Cid-Sánchez D, Flores-Balcázar CH, Celis-López MÁ, Gutiérrez-Aceves GA, Flores-Vázquez F, Moreno-Jiménez S. A Comprehensive Revision of Radiation Immunotherapy and the Abscopal Effect in Central Nervous System Metastases: Reassessing the Frontier. Curr Issues Mol Biol 2024; 46:11075-11085. [PMID: 39451538 PMCID: PMC11506806 DOI: 10.3390/cimb46100658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 10/26/2024] Open
Abstract
Seventy years ago, Robin Mole introduced the concept of the abscopal effect to describe a rare phenomenon. This occurs when local radiation triggers an immune-mediated reduction in tumors outside the treated area but within the same organism. Observing this effect has been linked to improved overall and progression-free survival in patients who experience it. While the abscopal effect was once considered rare, it is now being observed more frequently due to the combination of radiation with immunotherapy. As a result, more researchers are exploring this study area, which shows promise for excellent results. This review focuses explicitly on the immunological implications of activating the abscopal effect through ionizing radiation in the central nervous system and explores the potentially involved immunological pathways.
Collapse
Affiliation(s)
- Júlia Moscardini-Martelli
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Alejandro Rodríguez-Camacho
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
- Radiotherapy Service, National Cancer Institute, Mexico City 14080, Mexico; (F.J.L.-R.); (F.M.-M.); (D.C.-S.); (C.H.F.-B.)
| | - Jorge Alejandro Torres-Ríos
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Juan Marcos Meraz-Soto
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - José Guillermo Flores-Vázquez
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Laura Crystell Hernández-Sánchez
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Francisco Javier Lozano-Ruiz
- Radiotherapy Service, National Cancer Institute, Mexico City 14080, Mexico; (F.J.L.-R.); (F.M.-M.); (D.C.-S.); (C.H.F.-B.)
| | - Federico Maldonado-Magos
- Radiotherapy Service, National Cancer Institute, Mexico City 14080, Mexico; (F.J.L.-R.); (F.M.-M.); (D.C.-S.); (C.H.F.-B.)
| | - Dharely Cid-Sánchez
- Radiotherapy Service, National Cancer Institute, Mexico City 14080, Mexico; (F.J.L.-R.); (F.M.-M.); (D.C.-S.); (C.H.F.-B.)
| | | | - Miguel Ángel Celis-López
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | - Guillermo Axayacatl Gutiérrez-Aceves
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
| | | | - Sergio Moreno-Jiménez
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Mexico City 14269, Mexico; (J.M.-M.); (J.A.T.-R.); (J.M.M.-S.); (J.G.F.-V.); (L.C.H.-S.); (M.Á.C.-L.); (G.A.G.-A.); (S.M.-J.)
- American British Cowdray Medical Center, Neurological Center, Mexico City 01120, Mexico
| |
Collapse
|
3
|
Filograna A, De Tito S, Monte ML, Oliva R, Bruzzese F, Roca MS, Zannetti A, Greco A, Spano D, Ayala I, Liberti A, Petraccone L, Dathan N, Catara G, Schembri L, Colanzi A, Budillon A, Beccari AR, Del Vecchio P, Luini A, Corda D, Valente C. Identification and characterization of a new potent inhibitor targeting CtBP1/BARS in melanoma cells. J Exp Clin Cancer Res 2024; 43:137. [PMID: 38711119 PMCID: PMC11071220 DOI: 10.1186/s13046-024-03044-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 04/10/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND The C-terminal-binding protein 1/brefeldin A ADP-ribosylation substrate (CtBP1/BARS) acts both as an oncogenic transcriptional co-repressor and as a fission inducing protein required for membrane trafficking and Golgi complex partitioning during mitosis, hence for mitotic entry. CtBP1/BARS overexpression, in multiple cancers, has pro-tumorigenic functions regulating gene networks associated with "cancer hallmarks" and malignant behavior including: increased cell survival, proliferation, migration/invasion, epithelial-mesenchymal transition (EMT). Structurally, CtBP1/BARS belongs to the hydroxyacid-dehydrogenase family and possesses a NAD(H)-binding Rossmann fold, which, depending on ligands bound, controls the oligomerization of CtBP1/BARS and, in turn, its cellular functions. Here, we proposed to target the CtBP1/BARS Rossmann fold with small molecules as selective inhibitors of mitotic entry and pro-tumoral transcriptional activities. METHODS Structured-based screening of drug databases at different development stages was applied to discover novel ligands targeting the Rossmann fold. Among these identified ligands, N-(3,4-dichlorophenyl)-4-{[(4-nitrophenyl)carbamoyl]amino}benzenesulfonamide, called Comp.11, was selected for further analysis. Fluorescence spectroscopy, isothermal calorimetry, computational modelling and site-directed mutagenesis were employed to define the binding of Comp.11 to the Rossmann fold. Effects of Comp.11 on the oligomerization state, protein partners binding and pro-tumoral activities were evaluated by size-exclusion chromatography, pull-down, membrane transport and mitotic entry assays, Flow cytometry, quantitative real-time PCR, motility/invasion, and colony assays in A375MM and B16F10 melanoma cell lines. Effects of Comp.11 on tumor growth in vivo were analyzed in mouse tumor model. RESULTS We identify Comp.11 as a new, potent and selective inhibitor of CtBP1/BARS (but not CtBP2). Comp.11 directly binds to the CtBP1/BARS Rossmann fold affecting the oligomerization state of the protein (unlike other known CtBPs inhibitors), which, in turn, hinders interactions with relevant partners, resulting in the inhibition of both CtBP1/BARS cellular functions: i) membrane fission, with block of mitotic entry and cellular secretion; and ii) transcriptional pro-tumoral effects with significantly hampered proliferation, EMT, migration/invasion, and colony-forming capabilities. The combination of these effects impairs melanoma tumor growth in mouse models. CONCLUSIONS: This study identifies a potent and selective inhibitor of CtBP1/BARS active in cellular and melanoma animal models revealing new opportunities to study the role of CtBP1/BARS in tumor biology and to develop novel melanoma treatments.
Collapse
Affiliation(s)
- Angela Filograna
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Stefano De Tito
- Molecular Cell Biology of Autophagy, The Francis Crick Institute, London, UK. The Study Has Been Previously Performed at IEOS-CNR, Naples, Italy
| | - Matteo Lo Monte
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Rosario Oliva
- Department of Chemical Sciences, University of Naples Federico II, 80126, Naples, Italy
| | - Francesca Bruzzese
- Animal Facility Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Maria Serena Roca
- Experimental Pharmacology Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, Naples, 80131, Italy
| | - Antonella Zannetti
- Institute of Biostructures and Bioimaging (IBB), National Research Council (CNR), Naples, 80145, Italy
| | - Adelaide Greco
- Interdepartmental Service Center of Veterinary Radiology, University of Naples Federico II, 80137, Naples, Italy
| | - Daniela Spano
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Inmaculada Ayala
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Assunta Liberti
- National Research Council (CNR), Piazzale Aldo Moro, 700185, Rome, Italy
- Biology and Evolution of Marine Organisms (BEOM), Stazione Zoologica Anton Dohrn, Naples, Italy
| | - Luigi Petraccone
- Department of Chemical Sciences, University of Naples Federico II, 80126, Naples, Italy
| | - Nina Dathan
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Giuliana Catara
- Institute of Biochemistry and Cell Biology, National Research Council (CNR), 80131, Naples, Italy
| | - Laura Schembri
- National Research Council (CNR), Piazzale Aldo Moro, 700185, Rome, Italy
- Department of Pharmacy, University of Naples Federico II, 80131, Naples, Italy
| | - Antonino Colanzi
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Alfredo Budillon
- Scientific Directorate, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | | | - Pompea Del Vecchio
- Department of Chemical Sciences, University of Naples Federico II, 80126, Naples, Italy
| | - Alberto Luini
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy
| | - Daniela Corda
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy.
| | - Carmen Valente
- Institute of Experimental Endocrinology and Oncology "G. Salvatore"(IEOS), National Research Council (CNR), 80131, Naples, Italy.
- Present address: Dompé Farmaceutici S.P.A, L'Aquila, Italy.
| |
Collapse
|
4
|
Bashari N, Safaei Lari M, Darvishi A, Daroudi R. Cost-utility analysis of Pembrolizumab compared to other alternative immunotherapy and chemotherapy treatments for patients with advanced melanoma in Iran. Expert Rev Pharmacoecon Outcomes Res 2024; 24:273-284. [PMID: 37750606 DOI: 10.1080/14737167.2023.2263164] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/07/2023] [Indexed: 09/27/2023]
Abstract
OBJECTIVES Immunotherapy drugs like Pembrolizumab have shown significant improvements in treatment outcomes of advanced melanoma. This study aimed to evaluate the cost-utility of Pembrolizumab compared to other immunotherapy and chemotherapy drugs in the first-line treatment of advanced melanoma in Iran. METHODS A partitioned-survival model, based on data from a recent randomized phase 3 study (KEYNOTE-006) and recent meta-analysis, was used to divide Overall survival (OS) time into Progression-free survival (PFS) and post-progression survival for Pembrolizumab, Nivolumab, Ipilimumab, Dacarbazine, Temozolomide, Carboplatin, and Paclitaxel combination. Quality Life Years (QALY) and Incremental Cost-Effectiveness Ratio (ICER) were considered as the final outcome. RESULTS The ICER of Ipilimumab, Nivolumab, Nivolumab & Ipilimumab, and Pembrolizumab compared to Temozolomide was calculated as $40,365.53, $19,591.13, $24,578, and $47,324.2 per QALY, respectively. Scenario analysis demonstrated if the price of one vial of Nivolumab 100 is $90.51, each vial of Pembrolizumab is $119.20, and each vial of Ipilimumab is $101.54, they will be cost-effective in Iran. CONCLUSION None of the immunotherapy drugs studied were found to be cost-effective when considering the cost-effectiveness threshold of $3,532. Therefore, a cost reduction of more than 90% in the prices of immunotherapy drugs would be necessary for them to be considered cost-effective in Iran.
Collapse
Affiliation(s)
- Negin Bashari
- National Center for Health Insurance Research, Tehran, Iran
- Department of Health Management, Policy and Economics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Safaei Lari
- Department of Health Management, Policy and Economics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Darvishi
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Rajabali Daroudi
- National Center for Health Insurance Research, Tehran, Iran
- Department of Health Management, Policy and Economics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Chen JT, Zhou YW, Han TR, Wei JL, Qiu M. Perioperative immune checkpoint inhibition for colorectal cancer: recent advances and future directions. Front Immunol 2023; 14:1269341. [PMID: 38022667 PMCID: PMC10679411 DOI: 10.3389/fimmu.2023.1269341] [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: 07/29/2023] [Accepted: 10/25/2023] [Indexed: 12/01/2023] Open
Abstract
For colorectal cancer (CRC), surgical resection remains essential for achieving good prognoses. Unfortunately, numerous patients with locally advanced CRC and metastatic CRC failed to meet surgical indications or achieve pathological complete response after surgery. Perioperative therapy has been proven to effectively lower tumor staging and reduce recurrence and metastasis. Immune checkpoint inhibitors (ICIs) have shown unprecedented prolongation of survival time and satisfactory safety in patients with high microsatellite instability/deficient mismatch repair (MSI-H/dMMR), while the therapeutic effect obtained by patients with mismatch repair-proficient or microsatellite stable (pMMR/MSS) was considered minimal. However, recent studies found that certain CRC patients with dMMR/MSI-H presented intrinsic or acquired immune resistance, and pMMR/MSS CRC patients can also achieve better efficacy. Therefore, more predictors are required for screening patients with potential clinical benefits. Since the discovery of synergistic effects between immunotherapy, chemotherapy, and radiotherapy, different immunotherapy-based therapies have been applied to the perioperative therapy of CRC in an increasing number of research. This review comprehensively summarized the past and current progress of different combinations of immunotherapy in perioperative clinical trials for CRC, focusing on the efficacy and safety, and points out the direction for future development.
Collapse
Affiliation(s)
- Jiao-Ting Chen
- Department of Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yu-Wen Zhou
- Department of Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ting-Rui Han
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Jun-Lun Wei
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Meng Qiu
- Department of Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Yang F, Xu N. Letter Re: Response to checkpoint inhibition and targeted therapy in melanoma patients with concurrent haematological malignancies. Eur J Cancer 2023; 191:112982. [PMID: 37567797 DOI: 10.1016/j.ejca.2023.112982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 07/04/2023] [Indexed: 08/13/2023]
Affiliation(s)
- Fan Yang
- Department of Dermatology, Shengjing Hospital Affiliated to China Medical University, Liaoning, China
| | - Nan Xu
- Department of Plastic Surgery, The First Hospital of China Medical University, Liaoning, China.
| |
Collapse
|
7
|
Tagliaferri L, Lancellotta V, Fionda B, Mangoni M, Casà C, Di Stefani A, Pagliara MM, D’Aviero A, Schinzari G, Chiesa S, Mazzarella C, Manfrida S, Colloca GF, Marazzi F, Morganti AG, Blasi MA, Peris K, Tortora G, Valentini V. Immunotherapy and radiotherapy in melanoma: a multidisciplinary comprehensive review. Hum Vaccin Immunother 2022; 18:1903827. [PMID: 33847208 PMCID: PMC9122308 DOI: 10.1080/21645515.2021.1903827] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Melanoma is an extremely aggressive tumor and is considered to be an extremely immunogenic tumor because compared to other cancers it usually presents a well-expressed lymphoid infiltration. The aim of this paper is to perform a multidisciplinary comprehensive review of the evidence available about the combination of radiotherapy and immunotherapy for melanoma. Radiation, in fact, can increase tumor antigens visibility and promote priming of T cells but can also exert immunosuppressive action on tumor microenvironment. Combining radiotherapy with immunotherapy provides an opportunity to increase immunostimulatory potential of radiation. We therefore provide the latest clinical evidence about radiobiological rationale, radiotherapy techniques, timing, and role both in advanced and systemic disease (with a special focus on ocular melanoma and brain, liver, and bone metastases) with a particular attention also in geriatric patients. The combination of immunotherapy and radiotherapy seems to be a safe therapeutic option, supported by a clear biological rationale, even though the available data confirm that radiotherapy is employed more for metastatic than for non-metastatic disease. Such a combination shows promising results in terms of survival outcomes; however, further studies, hopefully prospective, are needed to confirm such evidence.
Collapse
Affiliation(s)
- Luca Tagliaferri
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Valentina Lancellotta
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Bruno Fionda
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
- CONTACT Bruno Fionda UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Largo Agostino Gemelli, 8, Roma00168, Italy
| | - Monica Mangoni
- Sezione di Radioterapia Oncologica, Dipartimento di Scienze Biomediche Sperimentali e Cliniche, Università di Firenze, Florence, Italy
| | - Calogero Casà
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Alessandro Di Stefani
- UOC Dermatologia, Dipartimento di Scienze mediche e chirurgiche, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Monica Maria Pagliara
- UOC Oncologia Oculare, Dipartimento di Scienze dell'Invecchiamento, neurologiche ortopediche e della testa collo, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Andrea D’Aviero
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Giovanni Schinzari
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
- Università Cattolica del Sacro Cuore, Roma, Italy
| | - Silvia Chiesa
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Ciro Mazzarella
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Stefania Manfrida
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Giuseppe Ferdinando Colloca
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Fabio Marazzi
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
| | - Alessio Giuseppe Morganti
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale Settore Scientifico Disciplinare, Università di Bologna, Bologna, Italy
| | - Maria Antonietta Blasi
- UOC Oncologia Oculare, Dipartimento di Scienze dell'Invecchiamento, neurologiche ortopediche e della testa collo, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
- Università Cattolica del Sacro Cuore, Roma, Italy
| | - Ketty Peris
- UOC Dermatologia, Dipartimento di Scienze mediche e chirurgiche, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
- Università Cattolica del Sacro Cuore, Roma, Italy
| | - Giampaolo Tortora
- UOC Oncologia Medica, Comprehensive Cancer Center, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
- Università Cattolica del Sacro Cuore, Roma, Italy
| | - Vincenzo Valentini
- UOC Radioterapia Oncologica, Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Roma, Italy
- Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
8
|
Role of Biomarkers in the Integrated Management of Melanoma. DISEASE MARKERS 2022; 2021:6238317. [PMID: 35003391 PMCID: PMC8739586 DOI: 10.1155/2021/6238317] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 12/13/2021] [Indexed: 12/21/2022]
Abstract
Melanoma, which is an aggressive skin cancer, is currently the fifth and seventh most common cancer in men and women, respectively. The American Cancer Society reported that approximately 106,110 new cases of melanoma were diagnosed in the United States in 2021, with 7,180 people dying from the disease. This information could facilitate the early detection of possible metastatic lesions and the development of novel therapeutic techniques for melanoma. Additionally, early detection of malignant melanoma remains an objective of melanoma research. Recently, melanoma treatment has substantially improved, given the availability of targeted treatments and immunotherapy. These developments have highlighted the significance of identifying biomarkers for prognosis and predicting therapy response. Biomarkers included tissue protein expression, circulating DNA detection, and genetic alterations in cancer cells. Improved diagnostic and prognostic biomarkers are becoming increasingly relevant in melanoma treatment, with the development of newer and more targeted treatments. Here, the author discusses the aspects of biomarkers in the real-time management of patients with melanoma.
Collapse
|
9
|
Pang H, Zhou Y, Wang J, Wu H, Cui C, Xiao Z. SKA3 overexpression predicts poor outcomes in skin cutaneous melanoma patients. Transl Oncol 2021; 15:101253. [PMID: 34737118 PMCID: PMC8571110 DOI: 10.1016/j.tranon.2021.101253] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/12/2021] [Accepted: 10/20/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Spindle and Kinetochore Associated Complex Subunit 3 (SKA3) is a part of the SKA complex, which plays a key role in cell mitosis. Studies have shown that SKA3 was associated with cancer progression. However, its role in skin cutaneous melanoma (SKCM) remains unclear. Here, we investigated the expression level and prognostic value of SKA3 in SKCM. METHODS Based on public databases, univariate and multivariate Cox regression analyses were used to investigate the different expression of SKA3 between SKCM and normal tissues. Then, the relationship between SKA3 expression level and prognosis was assessed. PPI network and functional enrichment analysis were performed. ESTIMATE and CIBERSORT were expected to evaluate the SKA3 expression and immune status. CCK8, wound healing, transwell assays and tumor xenograft trial were performed to detect the SKA3 function in cell viability, migration and invasion of the cell lines. RESULTS The SKA3 was highly expressed in SKCM tissues. SKA3 overexpression was associated with poor survival and immune status. SKA3 knockdown inhibited cell viability, migration and invasion of SKCM cells. CONCLUSION SKA3 is involved in the progression of SKCM and may serve as a new prognostic biomarker and therapeutic target.
Collapse
Affiliation(s)
- Hao Pang
- Department of Plastic Surgery, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Avenue, Harbin, Heilongjiang 150086, China
| | - Yongting Zhou
- Department of Plastic Surgery, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Avenue, Harbin, Heilongjiang 150086, China
| | - Jie Wang
- Department of Plastic Surgery, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Avenue, Harbin, Heilongjiang 150086, China
| | - Hao Wu
- Department of Plastic Surgery, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Avenue, Harbin, Heilongjiang 150086, China
| | - Chenyang Cui
- Department of Plastic Surgery, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Avenue, Harbin, Heilongjiang 150086, China
| | - Zhibo Xiao
- Department of Plastic Surgery, The Second Affiliated Hospital of Harbin Medical University, No. 246 Xuefu Avenue, Harbin, Heilongjiang 150086, China.
| |
Collapse
|
10
|
He Z, Xin Z, Peng Y, Zhao H, Fang X. Construction of competing endogenous RNA interaction network as prognostic markers in metastatic melanoma. PeerJ 2021; 9:e12143. [PMID: 34616613 PMCID: PMC8449535 DOI: 10.7717/peerj.12143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 08/19/2021] [Indexed: 11/20/2022] Open
Abstract
Malignant melanoma (MM) is a malignant tumor originating from melanocytes, with high aggressiveness, high metastasis and extremely poor prognosis. MM accounts for 4% of skin cancers and 80% of mortality, and the median survival of patients with metastatic melanoma is only about 6 months, with a five-year survival rate of less than 10%. In recent years, the incidence of melanoma has gradually increased and has become one of the serious diseases that endanger human health. Competitive endogenous RNA (ceRNA) is the main model of the mechanism by which long chain non-coding RNAs (lncRNAs) play a regulatory role in the disease. LncRNAs can act as a "sponge", competitively attracting small RNAs (micoRNAs; miRNAs), thus interfering with miRNA function, and affect the expression of target gene messenger RNAs (mRNAs), ultimately promoting tumorigenesis and progression. Bioinformatics analysis can identify potentially prognostic and therapeutically relevant differentially expressed genes in MM, finding lncRNAs, miRNAs and mRNAs that are interconnected through the ceRNA network, providing further insight into gene regulation and prognosis of metastatic melanoma. Weighted co-expression networks were used to identify lncRNA and mRNA modules associated with the metastatic phenotype, as well as the co-expression genes contained in the modules. A total of 17 lncRNAs, six miRNAs, and 11 mRNAs were used to construct a ceRNA interaction network that plays a regulatory role in metastatic melanoma patients. The prognostic risk model was used as a sorter to classify the survival prognosis of melanoma patients. Four groups of ceRNA interaction triplets were finally obtained, which miR-3662 might has potential implication for the treatment of metaststic melanoma patients, and futher experiments confirmed the regulating relationship and phenotype of this assumption. This study provides new targets to regulate metastatic process, predict metastatic potential and indicates that the miR-3662 can be used in the treatment of melanoma.
Collapse
Affiliation(s)
- Zan He
- Department of Dermatology, General Hospital of People's Liberation Army, Beijing, China.,Medical School of Chinese People's Liberation Army, Beijing, China
| | - Zijuan Xin
- Beijing Institute of Genomics/China National Center for Bioinformation, Chinese Academy of Science, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yongfei Peng
- Beijing Institute of Genomics/China National Center for Bioinformation, Chinese Academy of Science, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Hua Zhao
- Department of Dermatology, General Hospital of People's Liberation Army, Beijing, China.,Medical School of Chinese People's Liberation Army, Beijing, China
| | - Xiangdong Fang
- Beijing Institute of Genomics/China National Center for Bioinformation, Chinese Academy of Science, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
11
|
Light- and Melanin Nanoparticle-Induced Cytotoxicity in Metastatic Cancer Cells. Pharmaceutics 2021; 13:pharmaceutics13070965. [PMID: 34206894 PMCID: PMC8309021 DOI: 10.3390/pharmaceutics13070965] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/13/2021] [Accepted: 06/23/2021] [Indexed: 11/17/2022] Open
Abstract
Melanin nanoparticles are known to be biologically benign to human cells for a wide range of concentrations in a high glucose culture nutrition. Here, we show cytotoxic behavior at high nanoparticle and low glucose concentrations, as well as at low nanoparticle concentration under exposure to (nonionizing) visible radiation. To study these effects in detail, we developed highly monodispersed melanin nanoparticles (both uncoated and glucose-coated). In order to study the effect of significant cellular uptake of these nanoparticles, we employed three cancer cell lines: VM-M3, A375 (derived from melanoma), and HeLa, all known to exhibit strong macrophagic character, i.e., strong nanoparticle uptake through phagocytic ingestion. Our main observations are: (i) metastatic VM-M3 cancer cells massively ingest melanin nanoparticles (mNPs); (ii) the observed ingestion is enhanced by coating mNPs with glucose; (iii) after a certain level of mNP ingestion, the metastatic cancer cells studied here are observed to die—glucose coating appears to slow that process; (iv) cells that accumulate mNPs are much more susceptible to killing by laser illumination than cells that do not accumulate mNPs; and (v) non-metastatic VM-NM1 cancer cells also studied in this work do not ingest the mNPs, and remain unaffected after receiving identical optical energy levels and doses. Results of this study could lead to the development of a therapy for control of metastatic stages of cancer.
Collapse
|
12
|
Risk Factors for Relapse after Intentional Discontinuation of Immune Checkpoint Inhibitors in Melanoma Patients. J Immunother 2021; 44:239-241. [PMID: 34028389 DOI: 10.1097/cji.0000000000000375] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 04/12/2021] [Indexed: 11/26/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have tremendously changed the therapeutic landscape of melanoma since they are associated with a durable response, allowing for intentional discontinuation of therapy after complete or partial remission. However, a subset of patients develops a relapse after cessation of ICI treatment and may not respond to reinduction of ICIs. The aim of the present study was to identify risk factors for relapse after intentional discontinuation of ICI therapy. Patients with intentional discontinuation of ICI therapy for metastatic or unresectable melanoma from 5 German university hospitals were analyzed retrospectively. Clinicopathologic and follow-up data of 87 patients were collected and analyzed by univariate and multivariate Cox proportional-hazards models. The following parameters were associated with relapse after cessation of ICI treatment in the univariate Cox regression analysis: concurrent radiotherapy and ICI, best overall response, and presence of brain metastases. Duration of treatment, type of primary tumor, body mass index, programmed-death ligand 1 expression, and lactate dehydrogenase levels did not significantly influence the risk for relapse. In the multivariate analysis, partial remission [hazard ratio 4.217 (95% confidence interval: 1.424-12.49), P=0.009] and stable disease [3.327 (1.204-9.19), P=0.02] were associated with a significant decrease in progression-free survival compared with complete remission. Concurrent radiotherapy and ICI [3.619 (1.288-10.168), P=0.015] are additional independent risk factors for decreased progression-free survival upon ICI discontinuation, whereas the presence of brain metastasis did not reach statistical significance on multivariate analysis.
Collapse
|
13
|
Čelakovská J, Bukač J, Čáková L, Šimková M, Jandová E. Melanoma Incidence in Czech Republic, the Relation between Histology, Body Site of Melanoma, and Duration of Lesions. ACTA MEDICA (HRADEC KRÁLOVÉ) 2021; 63:1-9. [PMID: 32422110 DOI: 10.14712/18059694.2020.9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
AIM To evaluate the occurrence of melanoma in the period 1996-2017 in East Bohemia region in the Czech Republic. METHOD We studied the incidence of melanoma and the age of diagnosis (adjusted calculation) and the parameters such as histology, body site of lesions, the length of the duration of lesions in 2810 patients. RESULTS AND CONCLUSION No change in the occurrence of melanoma and in age of melanoma during this period was found. The difference between men and women was not confirmed in histology, but the difference between men and women was confirmed in the body site of lesion and in the length of duration of lesion. No relation between the length of duration of lesions from which melanoma had originated and its histology was confirmed. The relation was confirmed between histology and body site of melanoma. The relation between the body site and the length of duration of previous lesions was confirmed also. The increasing occurrence of melanoma on the trunk according to the duration of the previous lesions was confirmed.
Collapse
Affiliation(s)
- Jarmila Čelakovská
- Department of Dermatology and Venereology Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czech Republic.
| | - Josef Bukač
- Department of Medical Biophysics, Medical Faculty of Charles University, Hradec Králové, Czech Republic
| | - Lenka Čáková
- Department of Dermatology and Venereology Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czech Republic
| | - Marie Šimková
- Department of Dermatology and Venereology Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czech Republic
| | - Eva Jandová
- Department of Dermatology and Venereology Faculty Hospital and Medical Faculty of Charles University, Hradec Králové, Czech Republic
| |
Collapse
|
14
|
Boer FL, Ten Eikelder MLG, van Geloven N, Kapiteijn EH, Gaarenstroom KN, Hughes G, Nooij LS, Jozwiak M, Tjiong MY, de Hullu JMA, Galaal K, van Poelgeest MIE. Evaluation of treatment, prognostic factors, and survival in 198 vulvar melanoma patients: Implications for clinical practice. Gynecol Oncol 2021; 161:202-210. [PMID: 33514483 DOI: 10.1016/j.ygyno.2021.01.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 01/18/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To identify clinicopathological characteristics, treatment patterns, clinical outcomes and prognostic factors in patients with vulvar melanoma (VM). MATERIALS & METHODS This retrospective multicentre cohort study included 198 women with VM treated in eight cancer centres in the Netherlands and UK between 1990 and 2017. Clinicopathological features, treatment, recurrence, and survival data were collected. Overall and recurrence-free survival was estimated with the Kaplan-Meier method. Prognostic parameters were identified with multivariable Cox regression analysis. RESULTS The majority of patients (75.8%) had localized disease at diagnosis. VM was significantly associated with high-risk clinicopathological features, including age, tumour thickness, ulceration, positive resection margins and involved lymph nodes. Overall survival was 48% (95% CI 40-56%) and 31% (95% CI 23-39%) after 2 and 5 years respectively and did not improve in patients diagnosed after 2010 compared to patients diagnosed between 1990 and 2009. Recurrence occurred in 66.7% of patients, of which two-third was non-local. In multivariable analysis, age and tumour size were independent prognostic factors for worse survival. Prognostic factors for recurrence were tumour size and tumour type. Only the minority of patients were treated with immuno- or targeted therapy. CONCLUSION Our results show that even clinically early-stage VM is an aggressive disease associated with poor clinical outcome due to distant metastases. Further investigation into the genomic landscape and the immune microenvironment in VM may pave the way to novel therapies to improve clinical outcomes in these aggressive tumours. Clinical trials with immunotherapy or targeted therapy in patients with high-risk, advanced or metastatic disease are highly needed.
Collapse
Affiliation(s)
- Florine L Boer
- Department of Gynaecology, Leiden University Medical Centre, Leiden, the Netherlands.
| | | | - Nan van Geloven
- Department of Biomedical Data Sciences, Leiden University Medical Centre, the Netherlands
| | - Ellen H Kapiteijn
- Department of Medical Oncology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Katja N Gaarenstroom
- Department of Gynaecology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Geoff Hughes
- Department of Gynaecology, Derriford hospital NHS Trust, Plymouth, United Kingdom
| | - Linda S Nooij
- Department of Gynaecology Oncology, Centre for Gynaecologic Oncology, the Netherlands Cancer Institute, Antoni van Leeuwenhoek, the Netherlands
| | - Marta Jozwiak
- Department of Gynaecology Oncology, Erasmus MC Cancer Institute, Erasmus Medical Centre, the Netherlands
| | - Ming Y Tjiong
- Department of Gynaecology Oncology, Amsterdam University Medical Centre, the Netherlands
| | - Joanne M A de Hullu
- Department of Gynaecology Oncology, Radboud University Medical Centre, the Netherlands
| | - Khadra Galaal
- Department of Gynaecology, Royal Cornwall hospital NHS trust, Truro, United Kingdom
| | | |
Collapse
|
15
|
Li AL, Zhu YM, Gao LQ, Wei SY, Wang MT, Ma Q, Zheng YY, Li JH, Wang QF. Exploration of the Immune-Related Signatures and Immune Infiltration Analysis in Melanoma. Anal Cell Pathol (Amst) 2021; 2021:4743971. [PMID: 33511023 PMCID: PMC7826228 DOI: 10.1155/2021/4743971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 10/21/2020] [Indexed: 11/17/2022] Open
Abstract
In the present study, we aimed to investigate immune-related signatures and immune infiltration in melanoma. The transcriptome profiling and clinical data of melanoma were downloaded from The Cancer Genome Atlas database, and their matched normal samples were obtained from the Genotype-Tissue Expression database. After merging the genome expression data using Perl, the limma package was used for data normalization. We screened the differentially expressed genes (DEGs) and obtained immune signatures associated with melanoma by an immune-related signature list from the InnateDB database. Univariate Cox regression analysis was used to identify potential prognostic immune genes, and LASSO analysis was used to identify the hub genes. Next, based on the results of multivariate Cox regression analysis, we constructed a risk model for melanoma. We investigated the correlation between risk score and clinical characteristics and overall survival (OS) of patients. Based on the TIMER database, the association between selected immune signatures and immune cell distribution was evaluated. Next, the Wilcoxon rank-sum test was performed using CIBERSORT, which confirmed the differential distribution of immune-infiltrating cells between different risk groups. We obtained a list of 91 differentially expressed immune-related signatures. Functional enrichment analysis indicated that these immune-related DEGs participated in several areas of immune-related crosstalk, including cytokine-cytokine receptor interactions, JAK-STAT signaling pathway, chemokine signaling pathway, and Th17 cell differentiation pathway. A risk model was established based on multivariate Cox analysis results, and Kaplan-Meier analysis was performed. The Kruskal-Wallis test suggested that a high risk score indicated a poorer OS and correlated with higher American Joint Committee on Cancer-TNM (AJCC-TNM) stages and advanced pathological stages (P < 0.01). Furthermore, the association between hub immune signatures and immune cell distribution was evaluated in specific tumor samples. The Wilcoxon rank-sum test was used to estimate immune infiltration density in the two groups, and results showed that the high-risk group exhibited a lower infiltration density, and the dominant immune cells included M0 macrophages (P = 0.023) and activated mast cells (P = 0.005).
Collapse
Affiliation(s)
- Ai-lan Li
- Department of Dermatology, Dongying People's Hospital, Dongying 257091, China
| | - Yong-mei Zhu
- Department of Dermatology, Dongying People's Hospital, Dongying 257091, China
| | - Lai-qiang Gao
- Department of Dermatology, Dongying People's Hospital, Dongying 257091, China
| | - Shu-yue Wei
- Department of Dermatology, Dongying People's Hospital, Dongying 257091, China
| | - Ming-tao Wang
- Department of Dermatology, Dongying People's Hospital, Dongying 257091, China
| | - Qiang Ma
- Department of Dermatology, Dongying People's Hospital, Dongying 257091, China
| | - You-you Zheng
- Department of Dermatology, Dongying People's Hospital, Dongying 257091, China
| | - Jian-hua Li
- Department of Dermatology, Dongying People's Hospital, Dongying 257091, China
| | - Qing-feng Wang
- College of Integrated Chinese and Western Medicine, Liaoning University of traditional Chinese Medicine, Shenyang 110079, China
| |
Collapse
|
16
|
Zheng Q, Li J, Zhang H, Wang Y, Zhang S. Immune Checkpoint Inhibitors in Advanced Acral Melanoma: A Systematic Review. Front Oncol 2020; 10:602705. [PMID: 33344255 PMCID: PMC7744720 DOI: 10.3389/fonc.2020.602705] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 10/30/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Acral melanoma (AM) has different biological characteristics from cutaneous melanoma. Although systemic therapeutic strategies for advanced AM resemble those for advanced cutaneous melanoma, the evidence of the clinical use of immune checkpoint inhibitors (ICIs) for AM is still inadequate. We aimed to systematically analyze the therapeutic effects and safety profile of ICI treatments in advanced AM. METHODS This systematic review was conducted in line with a previously registered protocol. Three electronic databases, conference abstracts, clinical trial registers, and reference lists of included articles were searched for eligible studies. The primary outcomes were therapeutic effects, and the secondary outcomes were the safety profiles. RESULTS This systematic review included six studies investigating anti-CTLA-4 immunotherapy, 12 studies investigating anti-PD-1 immunotherapy, one study investigating the combination therapy of anti-CTLA-4 and anti-PD-1, and one study investigating anti-PD-1 immunotherapy in combination with radiotherapy. In most studies investigating ipilimumab, the anti-CTLA-4 antibody, the objective response rate ranged from 11.4 to 25%, the median progression-free survival ranged from 2.1 to 6.7 months, and the median overall survival was more than 7.16 months. For studies discussing anti-PD-1 immunotherapy with nivolumab, pembrolizumab, or JS001, the objective response rate ranged from 14 to 42.9%, the median progression-free survival ranged from 3.2 to 9.2 months, and the median overall survival was more than 14 months. The combination therapy of anti-CTLA-4 and anti-PD-1 immunotherapy showed better efficacy with an objective response rate of 42.9% than single-agent therapy. The retrospective study investigating the combination therapy of anti-PD-1 immunotherapy and radiation showed no overall response. Few outcomes regarding safety were reported in the included studies. CONCLUSIONS ICIs, especially anti-CTLA-4 monoclonal antibodies combined with anti-PD-1 antibodies, are effective systematic treatments in advanced AM. However, there remains a lack of high-level evidence to verify their efficacy and safety and support their clinical application.
Collapse
Affiliation(s)
- Qingyue Zheng
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Eight-year MD Program, Peking Union Medical College, Beijing, China
| | - Jiarui Li
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hanlin Zhang
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Eight-year MD Program, Peking Union Medical College, Beijing, China
| | - Yuanzhuo Wang
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Eight-year MD Program, Peking Union Medical College, Beijing, China
| | - Shu Zhang
- Department of Dermatology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
17
|
Urbanska AM, Khanin R, Alidori S, Wong S, Mello BP, Almeida BA, Chen F, Ma K, Turker MZ, Korontsvit T, Scheinberg DA, Zanzonico PB, Wiesner U, Bradbury MS, Quinn TP, McDevitt MR. A Genomic Profile of Local Immunity in the Melanoma Microenvironment Following Treatment with α Particle-Emitting Ultrasmall Silica Nanoparticles. Cancer Biother Radiopharm 2020; 35:459-473. [PMID: 32013538 PMCID: PMC7462037 DOI: 10.1089/cbr.2019.3150] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
An α particle-emitting nanodrug that is a potent and specific antitumor agent and also prompts significant remodeling of local immunity in the tumor microenvironment (TME) has been developed and may impact the treatment of melanoma. Biocompatible ultrasmall fluorescent core-shell silica nanoparticles (C' dots, diameter ∼6.0 nm) have been engineered to target the melanocortin-1 receptor expressed on melanoma through α melanocyte-stimulating hormone peptides attached to the C' dot surface. Actinium-225 is also bound to the nanoparticle to deliver a densely ionizing dose of high-energy α particles to cancer. Nanodrug pharmacokinetic properties are optimal for targeted radionuclide therapy as they exhibit rapid blood clearance, tumor-specific accumulation, minimal off-target localization, and renal elimination. Potent and specific tumor control, arising from the α particles, was observed in a syngeneic animal model of melanoma. Surprisingly, the C' dot component of this drug initiates a favorable pseudopathogenic response in the TME generating distinct changes in the fractions of naive and activated CD8 T cells, Th1 and regulatory T cells, immature dendritic cells, monocytes, MΦ and M1 macrophages, and activated natural killer cells. Concomitant upregulation of the inflammatory cytokine genome and adaptive immune pathways each describes a macrophage-initiated pseudoresponse to a viral-shaped pathogen. This study suggests that therapeutic α-particle irradiation of melanoma using ultrasmall functionalized core-shell silica nanoparticles potently kills tumor cells, and at the same time initiates a distinct immune response in the TME.
Collapse
MESH Headings
- Actinium/administration & dosage
- Actinium/pharmacokinetics
- Alpha Particles/therapeutic use
- Animals
- Cell Line, Tumor/transplantation
- Computational Biology
- Disease Models, Animal
- Dose-Response Relationship, Radiation
- Drug Carriers/chemistry
- Female
- Gene Expression Regulation, Neoplastic/immunology
- Gene Expression Regulation, Neoplastic/radiation effects
- Humans
- Immunity, Cellular/genetics
- Immunity, Cellular/radiation effects
- Male
- Maximum Tolerated Dose
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/radiotherapy
- Mice
- Molecular Targeted Therapy/methods
- Nanoparticles/chemistry
- RNA-Seq
- Radiopharmaceuticals/administration & dosage
- Radiopharmaceuticals/pharmacokinetics
- Receptor, Melanocortin, Type 1/antagonists & inhibitors
- Receptor, Melanocortin, Type 1/metabolism
- Silicon Dioxide/chemistry
- Skin Neoplasms/genetics
- Skin Neoplasms/immunology
- Skin Neoplasms/pathology
- Skin Neoplasms/radiotherapy
- Tissue Distribution
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
- Tumor Microenvironment/radiation effects
Collapse
Affiliation(s)
- Aleksandra M. Urbanska
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Raya Khanin
- Bioinformatics Core Computational Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Simone Alidori
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Sam Wong
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Chemistry, Hunter College, New York, New York, USA
| | - Barbara P. Mello
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Bryan Aristega Almeida
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Feng Chen
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Kai Ma
- Department of Materials Science & Engineering, Cornell University, Ithaca, New York, USA
| | - Melik Z. Turker
- Department of Materials Science & Engineering, Cornell University, Ithaca, New York, USA
| | - Tatyana Korontsvit
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - David A. Scheinberg
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Pharmacology, Weill Cornell Medicine College, New York, New York, USA
| | - Pat B. Zanzonico
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Ulrich Wiesner
- Department of Materials Science & Engineering, Cornell University, Ithaca, New York, USA
| | - Michelle S. Bradbury
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Molecular Pharmacology Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Radiology, Weill Cornell Medical College, New York, New York, USA
| | - Thomas P. Quinn
- Department of Biochemistry, University of Missouri, Columbia, Missouri, USA
- Harry S. Truman Veterans' Hospital, Columbia, Missouri, USA
| | - Michael R. McDevitt
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Department of Radiology, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
18
|
Stereotactic radiosurgery combined with anti-PD1 for the management of melanoma brain metastases: A retrospective study of safety and efficacy. Eur J Cancer 2020; 135:52-61. [PMID: 32535348 DOI: 10.1016/j.ejca.2020.04.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/07/2020] [Accepted: 04/07/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Brain metastases can be effectively treated with stereotactic radiosurgery (SRS). Immune checkpoint inhibitors are now pivotal in metastatic melanoma care, but some concerns have emerged regarding the safety of their combination with radiation therapy. METHODS We present a retrospective analysis of a cohort of patients treated by anti-PD1 and SRS as a sole modality of radiation therapy (no whole brain radiation therapy at any time) in a single institution. We included patients on anti-PD1 at the time of SRS or patients who started anti-PD1 within a maximum period of 3 months following SRS and were treated at least one year before the analysis. Clinical and serial imaging data were reviewed to determine the efficacy and the rate of adverse radiation effectss of the combination. RESULTS A total of 50 patients were included. SRS targeted 1, 2 to 3 and >3 brain metastases in 17, 16 and 17 patients, respectively. Two patients died before the first evaluation. Nine patients presented with an increase in peritumoral oedema, three with intracranial haemorrhage and one patient with both oedema and haemorrhage. Median follow-up was 38.89 months (interquartile range 24.43; 45.28). Median overall survival from SRS was 16.62 months with 1-, 2- and 3-year rates of 60%, 40% and 35%, respectively. Median brain-Progression Free Survival was 13.2 months with 1, 2 and 3-year rates of 62.1%, 49.7% and 49.7%, respectively. CONCLUSIONS This real-world cohort of patients treated with a homogeneous strategy combining upfront stereotactic radiosurgery and anti-PD1 shows remarkable survival rates and does not reveal unexpected toxicity.
Collapse
|
19
|
Li J, Kan H, Zhao L, Sun Z, Bai C. Immune checkpoint inhibitors in advanced or metastatic mucosal melanoma: a systematic review. Ther Adv Med Oncol 2020; 12:1758835920922028. [PMID: 32489431 PMCID: PMC7238311 DOI: 10.1177/1758835920922028] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/03/2020] [Indexed: 12/13/2022] Open
Abstract
Background Conventional cytotoxic chemotherapy offers minor benefit to patients with mucosal melanoma (MM). Although immune checkpoint inhibitors (ICIs) have become the preferred approach in patients with advanced or metastatic cutaneous melanoma, the evidence of their clinical use for MM is still limited. This systematic review aims to summarize the efficacy and safety of ICIs in advanced or metastatic MM. Methods We searched electronic databases, conference abstracts, clinical trial registers and reference lists for relevant studies. The primary outcomes included the overall response rate (ORR), median progression-free survival (PFS), median overall survival (OS), one-year PFS rate, and one-year OS rate. Results This review identified 13 studies assessing anti-CTLA-4 monotherapy, 22 studies assessing anti-PD-1 monotherapy, two studies assessing anti-CTLA-4 and anti-PD-1 combination therapy, one study assessing anti-PD-1 antibodies combined with axitinib, and three studies assessing anti-PD-1 antibodies combined with radiotherapy. For most patients who received ipilimumab monotherapy, the ORR ranged from 0% to 17%, the median PFS was less than 5 months, and the median OS was less than 10 months. For patients who received nivolumab or pembrolizumab monotherapy, most studies showed an ORR of more than 15% and a median OS of more than 11 months. The combined administration of anti-CTLA-4 and anti-PD-1 agents showed benefits over single-agent therapy with an ORR of more than 33.3%. In a phase Ib trial of toripalimab in combination with axitinib, approximately half of patients had complete or partial responses. Three retrospective studies that investigated anti-PD-1 antibodies combined with radiotherapy showed an ORR of more than 50%, which was higher than each single modality treatment. Conclusions Immune checkpoint inhibitors, especially anti-PD-1 monoclonal antibodies alone and in combination with anti-CTLA-4 monoclonal antibodies or other modalities, are promising treatment options for advanced or metastatic MM. However, high-level evidence is still needed to support the clinical application.
Collapse
Affiliation(s)
- Jiarui Li
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haoxuan Kan
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Zhao
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhao Sun
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunmei Bai
- Department of Medical Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuai Fu Yuan, Dongcheng District, Beijing 100032, China
| |
Collapse
|
20
|
Liu J, Xu C, Zhu J, Sivik J, Drabick JJ, Mackley HB. Identifying the Optimal Fractionation Schedules for Improved Response Rates and Survival in Patients with Metastatic Melanoma Treated with Ipilimumab and Radiotherapy. CURRENT CANCER THERAPY REVIEWS 2020. [DOI: 10.2174/2542584601666180326111906] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Objective:
There is a growing body of evidence that combining ipilimumab with higher
doses of radiotherapy may improve the response rates and survival in patients with metastatic melanoma
compared to lower doses of radiotherapy. However, the dose cutoff at which improved outcomes
are more likely to occur has not been properly identified.
Methods:
We conducted a retrospective analysis of 100 patients treated with ipilimumab and radiotherapy
for metastatic melanoma at a single institution from May 2011 to January 2017. Demographic,
clinical, and treatment factors, including the biological equivalent dose (BED) with an α/β
of 7, were recorded. Endpoints of interest included infield and global complete response (CR) after
the completion of radiation and ipilimumab based on the RECIST criteria (v1.1) and 12-month
overall survival (OS).
Results:
The BED cutoffs at which improved outcomes are more likely to occur are 46.5 Gy for infield
CR, 50.9 Gy for global CR, and 46.5 Gy for 12 month OS. The least aggressive fractionation
schedules used in this patient population that have a BED above the threshold for all 3 outcomes include
40 Gy in 20 fractions, 30 Gy in 6 fractions, and 24 Gy in 3 fractions.
Conclusion:
This hypothesis-generating study suggests that patients who cannot receive ablative intent
radiotherapy may be more likely to benefit from concurrent radiotherapy with ipilimumab if
their fractionation schedule has a BED above 46.5 - 50.9 Gy. Prospective trials evaluating this question
should be considered.
Collapse
Affiliation(s)
- Jason Liu
- The Penn State College of Medicine, Hershey, PA, United States
| | - Cong Xu
- The Division of Biostatistics at the Penn State Cancer Institute, Hershey, PA, United States
| | - Junjia Zhu
- The Division of Biostatistics at the Penn State Cancer Institute, Hershey, PA, United States
| | - Jeffrey Sivik
- The Division of Pharmacology at the Penn State Cancer Institute, Hershey, PA, United States
| | - Joseph J. Drabick
- The Division of Hematology/Oncology at the Penn State Cancer Institute, Hershey, PA, United States
| | - Heath B. Mackley
- The Division of Radiation Oncology at the Penn State Cancer Institute, Hershey, PA, United States
| |
Collapse
|
21
|
Wang LX, Wan C, Dong ZB, Wang BH, Liu HY, Li Y. Integrative Analysis of Long Noncoding RNA (lncRNA), microRNA (miRNA) and mRNA Expression and Construction of a Competing Endogenous RNA (ceRNA) Network in Metastatic Melanoma. Med Sci Monit 2019; 25:2896-2907. [PMID: 31004080 PMCID: PMC6487673 DOI: 10.12659/msm.913881] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Worldwide, metastatic melanoma of the skin has an aggressive course with high morbidity and mortality. Therefore, an increased understanding of the pathogenesis of metastatic melanoma has gained increasing attention, including the role of epigenetic modification and competing endogenous RNA (ceRNA). This study aimed to used bioinformatics data to undertake an integrative analysis of long noncoding RNA (lncRNA), microRNA (miRNA) and mRNA expression to construct a ceRNA network in metastatic melanoma. Data from the Cancer Genome Atlas (TCGA), the Gene Ontology (GO) database, and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway were analyzed. There were 471 cases that included 103 primary solid tumors and 368 cases of metastatic melanoma that included transcriptome sequencing data (including lncRNA and mRNA); 452 cases had miRNA sequencing data. Analysis of chip data identified 85 6 mRNAs, 67 miRNAs, and 250 lncRNAs that were differentially expressed in cases of metastatic melanoma, of which 25 miRNAs, 18 lncRNAs, and 18 mRNAs participated in the formation of ceRNAs. Survival analysis identified seven differentially expressed mRNAs, five differentially expressed miRNAs (miRNA-29c, miRNA-100, miR-142-3p, miR-150, miR-516a-2), and six differentially expressed lncRNAs (AC068594.1, C7orf71, FAM41C, GPC5-AS1, MUC19, LINC00402) that were correlated with survival time in patients with metastatic melanoma. Bioinformatics data and integrative analysis identified lncRNA, miRNA, and mRNA expression to construct a ceRNA and patient survival network in metastatic melanoma. These findings support the need for further studies on the mechanisms involved in the regulation of metastatic melanoma by ceRNAs.
Collapse
Affiliation(s)
- Li-Xin Wang
- Department of Dermatology, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| | - Chuan Wan
- Department of Dermatology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (mainland)
| | - Zheng-Bang Dong
- Department of Dermatology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, Jiangsu, China (mainland)
| | - Bai-He Wang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China (mainland)
| | - Hong-Ye Liu
- Department of Dermatology, The First Affiliated Hospital of Shanxi Medical University, Taiyuan, Shanxi, China (mainland)
| | - Yang Li
- Department of Dermatology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, Shandong, China (mainland)
| |
Collapse
|
22
|
Liu H, Yang L, Qi M, Zhang J. NFAT1 enhances the effects of tumor-associated macrophages on promoting malignant melanoma growth and metastasis. Biosci Rep 2018; 38:BSR20181604. [PMID: 30459241 PMCID: PMC6435508 DOI: 10.1042/bsr20181604] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/31/2018] [Accepted: 11/12/2018] [Indexed: 12/24/2022] Open
Abstract
Tumor-associated macrophages (TAMs) play substantial roles in tumor growth, invasion, and metastasis. Nuclear factor of activated T cell (NFAT1) has been shown to promote melanoma growth and metastasis in vivo We herein aim to investigate whether NFAT1 is capable to promote melanoma growth and metastasis by influencing TAM properties. Melanoma-conditioned TAMs were obtained from human monocytes after incubation with conditioned medium from A375 cell culture. The phenotype of the macrophages was detected. Cell proliferation, migration, and invasion were evaluated. Human malignant melanoma tissues exhibited increased CD68+-macrophage infiltration and NFAT1 expression compared with the normal pigmented nevus tissues. Melanoma-conditioned TAMs displayed M2-like phenotype. Melanoma-conditioned TAMs also promoted proliferation, migration, and invasion of human malignant melanoma cell lines A375 and WM451. Furthermore, NFAT1 expression in TAMs was significantly increased compared with the M0 group. NFAT1 overexpression significantly strengthened the melanoma-conditioned TAM-mediated promotion of cell migration and invasion in A375 and WM451 cells, whereas NFAT1 knockdown exerted the opposite effects. Moreover, NFAT1 overexpression in melanoma-conditioned TAMs promoted CD68+-macrophage infiltration, tumor growth, and metastasis in vivo NFAT1 may play a critical role in enhancing the TAM-mediated promotion of growth and metastasis in malignant melanoma.
Collapse
Affiliation(s)
- Hao Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, P.R. China
| | - Liping Yang
- College of Life Sciences, Hunan Normal University, Changsha, Hunan Province 410006, P.R. China
| | - Min Qi
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, P.R. China
| | - Jianglin Zhang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan Province 410008, P.R. China
| |
Collapse
|
23
|
Arcangeli S, Jereczek-Fossa BA, Alongi F, Aristei C, Becherini C, Belgioia L, Buglione M, Caravatta L, D'Angelillo RM, Filippi AR, Fiore M, Genovesi D, Greco C, Livi L, Magrini SM, Marvaso G, Mazzola R, Meattini I, Merlotti A, Palumbo I, Pergolizzi S, Ramella S, Ricardi U, Russi E, Trovò M, Sindoni A, Valentini V, Corvò R. Combination of novel systemic agents and radiotherapy for solid tumors - part I: An AIRO (Italian association of radiotherapy and clinical oncology) overview focused on treatment efficacy. Crit Rev Oncol Hematol 2018; 134:87-103. [PMID: 30658886 DOI: 10.1016/j.critrevonc.2018.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 11/13/2018] [Accepted: 11/20/2018] [Indexed: 02/07/2023] Open
Abstract
Over the past century, technologic advances have promoted the evolution of radiation therapy into a precise treatment modality allowing for the maximal administration of dose to tumors while sparing normal tissues. In parallel with this technological maturation, the rapid expansion in understanding the basic biology and heterogeneity of cancer has led to the development of several compounds that target specific pathways. Many of them are in advanced steps of clinical development for combination treatments with radiotherapy, and can be incorporated into radiation oncology practice for a personalized approach to maximize the therapeutic gain. This review describes the rationale for combining novel agents with radiation, and provides an overview of the current landscape focused on treatment efficacy.
Collapse
Affiliation(s)
- Stefano Arcangeli
- Department of Radiation Oncology, Policlinico S. Gerardo and University of Milan "Bicocca", Milan, Italy.
| | | | - Filippo Alongi
- Department of Radiation Oncology, Sacro Cuore Don Calabria Cancer Care Center, Negrar-Verona, and University of Brescia, Brescia, Italy
| | - Cynthia Aristei
- Radiation Oncology Section, Department of Surgical and Biomedical Science, University of Perugia, Perugia General Hospital, Perugia, Italy
| | - Carlotta Becherini
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Firenze, Italy
| | - Liliana Belgioia
- Department of Radiation Oncology, Ospedale Policlinico San Martino and University of Genoa, Genoa, Italy
| | - Michela Buglione
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Luciana Caravatta
- Department of Radiation Oncology, SS. Annunziata Hospital, G. D'Annunzio University of Chieti, Chieti, Italy
| | | | | | - Michele Fiore
- Radiotherapy Unit, Campus Bio-Medico University, Rome, Italy
| | - Domenico Genovesi
- Department of Radiation Oncology, SS. Annunziata Hospital, G. D'Annunzio University of Chieti, Chieti, Italy
| | - Carlo Greco
- Radiotherapy Unit, Campus Bio-Medico University, Rome, Italy
| | - Lorenzo Livi
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Firenze, Italy
| | - Stefano Maria Magrini
- Department of Radiation Oncology, University and Spedali Civili Hospital, Brescia, Italy
| | - Giulia Marvaso
- Deparment of Radiation Oncology of IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Rosario Mazzola
- Department of Radiation Oncology, Sacro Cuore Don Calabria Cancer Care Center, Negrar-Verona, and University of Brescia, Brescia, Italy
| | - Icro Meattini
- Radiotherapy Unit, Department of Experimental and Clinical Biomedical Sciences, University of Florence, Firenze, Italy
| | - Anna Merlotti
- Department of Radiation Oncology, S. Croce and Carle Teaching Hospital, Cuneo, Italy
| | - Isabella Palumbo
- Radiation Oncology Section, Department of Surgical and Biomedical Science, University of Perugia, Perugia General Hospital, Perugia, Italy
| | - Stefano Pergolizzi
- Department of Biomedical Sciences and Morphological and Functional Images, University of Messina, Italy
| | - Sara Ramella
- Radiotherapy Unit, Campus Bio-Medico University, Rome, Italy
| | | | - Elvio Russi
- Department of Radiation Oncology, S. Croce and Carle Teaching Hospital, Cuneo, Italy
| | - Marco Trovò
- Department of Radiation Oncology, Azienda Sanitaria Universitaria Integrata of Udine, Udine, Italy
| | - Alessandro Sindoni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Vincenzo Valentini
- Gemelli Advanced Radiation Therapy Center, Fondazione Policlinico Universitario "A. Gemelli", Catholic University of Sacred Heart, Rome, Italy
| | - Renzo Corvò
- Department of Radiation Oncology, Ospedale Policlinico San Martino and University of Genoa, Genoa, Italy
| |
Collapse
|
24
|
Zhao B, Zhao H, Zhao J. Impact of clinicopathological characteristics on survival in patients treated with immune checkpoint inhibitors for metastatic melanoma. Int J Cancer 2018; 144:169-177. [PMID: 30121946 DOI: 10.1002/ijc.31813] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/01/2018] [Accepted: 08/09/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Bin Zhao
- The Second Affiliated Hospital & Yuying Children's Hospital; Wenzhou Medical University; Wenzhou China
| | - Hong Zhao
- The Third Affiliated Hospital of Harbin Medical University; Harbin China
| | - Jiaxin Zhao
- The Fourth Affiliated Hospital of Harbin Medical University; Harbin China
| |
Collapse
|
25
|
Combined radiotherapy with nivolumab for extracranial metastatic malignant melanoma. Jpn J Radiol 2018; 36:712-718. [PMID: 30206802 DOI: 10.1007/s11604-018-0774-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/05/2018] [Indexed: 12/13/2022]
Abstract
PURPOSE We retrospectively evaluated the tumor regression after radiotherapy in combination with the immune checkpoint inhibitor nivolumab for metastatic melanoma. MATERIALS AND METHODS We evaluated the extracranial metastatic melanoma lesions to which concomitant radiotherapy with nivolumab was administered from June 2015 to February 2017. Tumor volume and maximum diameter were measured at the time of pre-radiotherapy and best response, and the tumor reduction rate was assessed in two ways that our hospital adopts: tumor volume and diameter. RESULTS Seven lesions in five patients were evaluated. The median time from the start of nivolumab treatment to the start of radiotherapy was 5 months (range 0-22 months). The objective response rate was 85.7% in the evaluation by tumor volume and 42.9% by maximum diameter of the tumor. The objective complete response rate was 28.6% in evaluation by tumor volume and 14.3% by maximum dia. The 1-year tumor control rate was 62.5%. The 1- and 2-year overall survival rate after nivolumab treatment were 75% and 50%, respectively. Two patients who obtained a complete response had presented with vitiligo. CONCLUSION The combination of radiotherapy and nivolumab treatment produced favorable responses. Vitiligo may be correlated with a good response to concomitant radiotherapy with nivolumab.
Collapse
|
26
|
Buttigliero C, Allis S, Tucci M, Zichi C, Leone G, Di Stefano RF, Ruo Redda MG, Ricardi U, Scagliotti GV, Di Maio M, Filippi AR. Role of radiotherapy in improving activity of immune-modulating drugs in advanced renal cancer: Biological rationale and clinical evidences. Cancer Treat Rev 2018; 69:215-223. [PMID: 30096699 DOI: 10.1016/j.ctrv.2018.07.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 07/17/2018] [Accepted: 07/19/2018] [Indexed: 12/17/2022]
Abstract
In the last few years, immune checkpoint inhibitors have been extensively investigated in renal cell carcinoma and led to remarkable results. Radiation therapy may increase the activity of immune modulating agents through different mechanisms, priming the immune system, recruiting immune cells to the tumor environment, and altering the immunosuppressive effects of the tumor microenvironment. Preclinical studies reported increased loco-regional control when radiation is combined with immune-checkpoint blockade. Moreover, increased systemic disease control has been demonstrated when local radiation is combined with both anti-CTLA-4 and anti-PD-1/PD-L1 inhibitors. Actually, several trials are ongoing testing the activity of radiation therapy in combination with different immune-modulating agents for the treatment of metastatic renal cell carcinoma. The aim of this paper is to focus on the biological rationale of adding radiation therapy to immune-modulating agents in renal cell carcinoma and to review the currently available clinical evidence about the combination of radiotherapy and immunotherapy.
Collapse
Affiliation(s)
- Consuelo Buttigliero
- Division of Medical Oncology, San Luigi Gonzaga Hospital, Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Simona Allis
- Radiation Oncology, San Luigi Gonzaga Hospital, Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Marcello Tucci
- Division of Medical Oncology, San Luigi Gonzaga Hospital, Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy.
| | - Clizia Zichi
- Division of Medical Oncology, San Luigi Gonzaga Hospital, Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Gianmarco Leone
- Division of Medical Oncology, San Luigi Gonzaga Hospital, Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Rosario Francesco Di Stefano
- Division of Medical Oncology, San Luigi Gonzaga Hospital, Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Maria Grazia Ruo Redda
- Radiation Oncology, Ordine Mauriziano Hospital, Department of Oncology, University of Turin, Via Magellano 1, 10028 Turin, Italy
| | - Umberto Ricardi
- Radiation Oncology, Città della Salute e della Scienza di Torino, Department of Oncology, University of Turin, Via Genova 3, 10126 Turin, Italy
| | - Giorgio Vittorio Scagliotti
- Division of Medical Oncology, San Luigi Gonzaga Hospital, Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Massimo Di Maio
- Division of Medical Oncology, Ordine Mauriziano Hospital, Department of Oncology, University of Turin, Via Magellano 1, 10028 Turin, Italy
| | - Andrea Riccardo Filippi
- Radiation Oncology, San Luigi Gonzaga Hospital, Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| |
Collapse
|
27
|
Gabani P, Robinson CG, Ansstas G, Johanns TM, Huang J. Use of extracranial radiation therapy in metastatic melanoma patients receiving immunotherapy. Radiother Oncol 2018. [DOI: 10.1016/j.radonc.2018.02.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
28
|
Filippi AR, Di Muzio J, Badellino S, Mantovani C, Ricardi U. Locally-advanced non-small cell lung cancer: shall immunotherapy be a new chance? J Thorac Dis 2018; 10:S1461-S1467. [PMID: 29951297 DOI: 10.21037/jtd.2017.12.53] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Locally advanced non-small cell lung cancer (NSCLC) represents approximately one third of presentations at diagnosis. Most patients are judged non-surgical due to disease extension, and chemo-radiotherapy still represents the standard therapeutic option, with unsatisfactory results in terms of overall survival (OS) despite advances in staging and radiation therapy planning and delivery. Immunotherapy, and in particular immune-checkpoint inhibitors targeting the PD-1/PD-L1 axis, gained wide popularity for NSCLC in light of the positive findings of several trials in metastatic disease. Stage III unresectable NSCLC is a remarkably interesting setting for the combined use of chemo-radiation and immunotherapy, also considering the multiple experimental evidences in favor of a synergistic effect between radiation and immune checkpoint inhibitors, with the potential of enhancing immuno-modulating effects and overcoming resistance. We here summarized the biological rationale and the initial clinical experiences testing for this combination, and we briefly discussed ongoing trials and future options in this field.
Collapse
Affiliation(s)
| | | | - Serena Badellino
- Radiation Oncology Department, Città della Salute e della Scienza University Hospital, Torino, Italy
| | - Cristina Mantovani
- Radiation Oncology Department, Città della Salute e della Scienza University Hospital, Torino, Italy
| | | |
Collapse
|
29
|
Khan M, Lin J, Liao G, Tian Y, Liang Y, Li R, Liu M, Yuan Y. SRS in Combination With Ipilimumab: A Promising New Dimension for Treating Melanoma Brain Metastases. Technol Cancer Res Treat 2018; 17:1533033818798792. [PMID: 30213236 PMCID: PMC6137552 DOI: 10.1177/1533033818798792] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 05/10/2018] [Accepted: 08/01/2018] [Indexed: 01/08/2023] Open
Abstract
Stereotactic radiosurgery provides effective local control, but high recurrence rate are observed while ipilimumab have shown promising improvements in survival in the treatment of melanoma brain metastases. This meta-analysis was done to review the clinical evidence regarding the combination of stereotactic radiosurgery and ipilimumab in the treatment of brain metastases from melanoma. Comprehensive research of the electronic databases (PubMed and Cochrane Library) was carried out in April 2017. Different combination of MESH headings and words were used. Review Manager was used to analyze the outcome data of interest. According to heterogeneity, fixed effects model or random effects model was adapted. Six retrospective studies comparing stereotactic radiosurgery plus ipilimumab with stereotactic radiosurgery alone were found. Total of 411 participants were included in this meta-analysis. Of that, 128 patients had received stereotactic radiosurgery + ipilimumab, while 283 patients had received stereotactic radiosurgery only. Stereotactic radiosurgery plus ipilimumab significantly improved survival when compared to stereotactic radiosurgery alone (hazard ratio: 0.74 [95% confidence interval: 0.56-0.99, P = .04]), with no significant increase in the incidence of adverse events (odds ratio 0.57 [95% confidence interval: 0.28-1.17, P = .12]). Stereotactic radiosurgery with ipilimumab is safe and effective treatment option and can be recommended for the treatment of brain metastases in patients with melanoma.
Collapse
Affiliation(s)
- Muhammad Khan
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute
of Guangzhou Medical University, Guangzhou, Guangdong Province, People’s Republic of
China
| | - Jie Lin
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute
of Guangzhou Medical University, Guangzhou, Guangdong Province, People’s Republic of
China
| | - Guixiang Liao
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute
of Guangzhou Medical University, Guangzhou, Guangdong Province, People’s Republic of
China
| | - Yunhong Tian
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute
of Guangzhou Medical University, Guangzhou, Guangdong Province, People’s Republic of
China
| | - Yingying Liang
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute
of Guangzhou Medical University, Guangzhou, Guangdong Province, People’s Republic of
China
| | - Rong Li
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute
of Guangzhou Medical University, Guangzhou, Guangdong Province, People’s Republic of
China
| | - Mengzhong Liu
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute
of Guangzhou Medical University, Guangzhou, Guangdong Province, People’s Republic of
China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Sun
Yat-sen Medical University, Guangzhou, Guangdong Province, People’s Republic of China
| | - Yawei Yuan
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute
of Guangzhou Medical University, Guangzhou, Guangdong Province, People’s Republic of
China
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Sun
Yat-sen Medical University, Guangzhou, Guangdong Province, People’s Republic of China
| |
Collapse
|
30
|
Stereotactic radiosurgery of early melanoma brain metastases after initiation of anti-CTLA-4 treatment is associated with improved intracranial control. Radiother Oncol 2017; 125:80-88. [PMID: 28916225 DOI: 10.1016/j.radonc.2017.08.009] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 07/19/2017] [Accepted: 08/05/2017] [Indexed: 01/20/2023]
Abstract
BACKGROUND Numerous studies suggest that radiation can boost antitumor immune response by stimulating release of tumor-specific antigens. However, the optimal timing between radiotherapy and immune checkpoint blockade to achieve potentially synergistic benefits is unclear. MATERIAL AND METHODS Multi-institutional retrospective analysis was conducted of ninety-nine metastatic melanoma patients from 2007 to 2014 treated with ipilimumab who later received stereotactic radiosurgery (SRS) for new brain metastases that developed after starting immunotherapy. All patients had complete blood count acquired before SRS. Primary outcomes were intracranial disease control and overall survival (OS). RESULTS The median follow-up time was 15.5months. In the MD Anderson cohort, patients who received SRS after 5.5months (n=20) of their last dose of ipilimumab had significantly worse intracranial control than patients who received SRS within 5.5months (n=51) (median 3.63 vs. 8.09months; hazard ratio [HR] 2.07, 95% confidence interval [CI] 1.03-4.16, p=0.041). OS was not different between the two arms. The improvement in intracranial control was confirmed in an independent validation cohort of 28 patients treated at Yale-New Haven Hospital. Circulating absolute lymphocyte count before SRS predicted for treatment response as those with baseline counts >1000/µL had reduced risk of intracranial recurrence compared with those with ≤1000/µL (HR 0.46, 95% CI 0.0.23-0.94, p=0.03). CONCLUSIONS In this multi-institutional study, patients who received SRS for new brain metastases within 5.5months after ipilimumab therapy had better intracranial disease control than those who received SRS later. Moreover, higher circulating lymphocyte count was associated with improved intracranial disease control.
Collapse
|
31
|
Rodemann HP, Datta NR, Bodis S. Molecular radiation biology/oncology and its impact on preclinical and clinical research in radiotherapy. Radiother Oncol 2017; 124:339-343. [PMID: 28888706 DOI: 10.1016/j.radonc.2017.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 08/22/2017] [Indexed: 11/20/2022]
Affiliation(s)
- H Peter Rodemann
- Division of Radiation Biology & Molecular Environmental Research, Dept. of Radiation Oncology, University of Tübingen, German Cancer Consortium (DKTK), partner site Tübingen, and German Cancer Research Center (DKFZ) Heidelberg, Germany.
| | - Niloy Ranjan Datta
- Center of Radiation Oncology KSA-KSB, Kantonsspital Aarau and University of Zurich, Switzerland
| | - Stephan Bodis
- Center of Radiation Oncology KSA-KSB, Kantonsspital Aarau and University of Zurich, Switzerland
| |
Collapse
|
32
|
Werner LR, Kler JS, Gressett MM, Riegert M, Werner LK, Heinze CM, Kern JG, Abbariki M, Erbe AK, Patel RB, Sriramaneni RN, Harari PM, Morris ZS. Transcriptional-mediated effects of radiation on the expression of immune susceptibility markers in melanoma. Radiother Oncol 2017; 124:418-426. [PMID: 28893414 DOI: 10.1016/j.radonc.2017.08.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/12/2017] [Accepted: 08/20/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND PURPOSE We recently reported a time-sensitive, cooperative, anti-tumor effect elicited by radiation (RT) and intra-tumoral-immunocytokine injection in vivo. We hypothesized that RT triggers transcriptional-mediated changes in tumor expression of immune susceptibility markers at delayed time points, which may explain these previously observed time-dependent effects. MATERIALS AND METHODS We examined the time course of changes in expression of immune susceptibility markers following in vitro or in vivo RT in B78 murine melanoma and A375 human melanoma using flow cytometry, immunoblotting, and qPCR. RESULTS Flow cytometry and immunoblot revealed time-dependent increases in expression of death receptors and T cell co-stimulatory/co-inhibitory ligands following RT in murine and human melanoma. Using high-throughput qPCR, we observed comparable time courses of RT-induced transcriptional upregulation for multiple immune susceptibility markers. We confirmed analogous changes in B78 tumors irradiated in vivo. We observed upregulated expression of DNA damage response markers days prior to changes in immune markers, whereas phosphorylation of the STAT1 transcription factor occurred concurrently with changes following RT. CONCLUSION This study highlights time-dependent, transcription-mediated changes in tumor immune susceptibility marker expression following RT. These findings may help in the design of strategies to optimize sequencing of RT and immunotherapy in translational and clinical studies.
Collapse
Affiliation(s)
- Lauryn R Werner
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, United States.
| | - Jasdeep S Kler
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, United States.
| | - Monica M Gressett
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, United States.
| | - Maureen Riegert
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, United States.
| | - Lindsey K Werner
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, United States.
| | - Clinton M Heinze
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, United States.
| | - Joseph G Kern
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, United States.
| | - Mahyar Abbariki
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, United States.
| | - Amy K Erbe
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, United States.
| | - Ravi B Patel
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, United States.
| | - Raghava N Sriramaneni
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, United States.
| | - Paul M Harari
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, United States.
| | - Zachary S Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, United States.
| |
Collapse
|
33
|
Radiation and PD-1 inhibition: Favorable outcomes after brain-directed radiation. Radiother Oncol 2017; 124:98-103. [DOI: 10.1016/j.radonc.2017.06.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 05/12/2017] [Accepted: 06/05/2017] [Indexed: 01/19/2023]
|
34
|
Ahmed KA, Kim S, Arrington J, Naghavi AO, Dilling TJ, Creelan BC, Antonia SJ, Caudell JJ, Harrison LB, Sahebjam S, Gray JE, Etame AB, Johnstone PA, Yu M, Perez BA. Outcomes targeting the PD-1/PD-L1 axis in conjunction with stereotactic radiation for patients with non-small cell lung cancer brain metastases. J Neurooncol 2017; 133:331-338. [DOI: 10.1007/s11060-017-2437-5] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 04/15/2017] [Indexed: 01/23/2023]
|
35
|
Kroeze SGC, Fritz C, Hoyer M, Lo SS, Ricardi U, Sahgal A, Stahel R, Stupp R, Guckenberger M. Toxicity of concurrent stereotactic radiotherapy and targeted therapy or immunotherapy: A systematic review. Cancer Treat Rev 2016; 53:25-37. [PMID: 28056412 DOI: 10.1016/j.ctrv.2016.11.013] [Citation(s) in RCA: 136] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 11/26/2016] [Indexed: 10/20/2022]
Abstract
BACKGROUND AND PURPOSE Both stereotactic radiotherapy (SRT) and immune- or targeted therapy play an increasingly important role in personalized treatment of metastatic disease. Concurrent application of both therapies is rapidly expanding in daily clinical practice. In this systematic review we summarize severe toxicity observed after concurrent treatment. MATERIAL AND METHODS PubMed and EMBASE databases were searched for English literature published up to April 2016 using keywords "radiosurgery", "local ablative therapy", "gamma knife" and "stereotactic", combined with "bevacizumab", "cetuximab", "crizotinib", "erlotinib", "gefitinib", "ipilimumab", "lapatinib", "sorafenib", "sunitinib", "trastuzumab", "vemurafenib", "PLX4032", "panitumumab", "nivolumab", "pembrolizumab", "alectinib", "ceritinib", "dabrafenib", "trametinib", "BRAF", "TKI", "MEK", "PD1", "EGFR", "CTLA-4" or "ALK". Studies performing SRT during or within 30days of targeted/immunotherapy, reporting severe (⩾Grade 3) toxicity were included. RESULTS Concurrent treatment is mostly well tolerated in cranial SRT, but high rates of severe toxicity were observed for the combination with BRAF-inhibitors. The relatively scarce literature on extra-cranial SRT shows a potential risk of increased toxicity when SRT is combined with EGFR-targeting tyrosine kinase inhibitors and bevacizumab, which was not observed for cranial SRT. CONCLUSIONS This review gives a best-possible overview of current knowledge and its limitations and underlines the need for a timely generation of stronger evidence in this rapidly expanding field.
Collapse
Affiliation(s)
- Stephanie G C Kroeze
- Department of Radiation Oncology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland.
| | - Corinna Fritz
- Department of Radiation Oncology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Morten Hoyer
- Danish Center for Particle Therapy, Aarhus University, Palle Juul-Jensens Boulevard, 8200 Aarhus, Denmark
| | - Simon S Lo
- Department of Radiation Oncology, University of Washington School of Medicine, 1959 N.E. Pacific Street, Box 356043, Seattle, USA
| | - Umberto Ricardi
- Department of Oncology, University of Turin, Regione Gonzole 10, 10043 Orbassano, Italy
| | - Arjun Sahgal
- Department of Radiation Oncology, University of Toronto, 27 King's College Circle Toronto, Ontario M5S 1A1, Canada
| | - Rolf Stahel
- Department of Oncology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Roger Stupp
- Department of Oncology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Matthias Guckenberger
- Department of Radiation Oncology, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| |
Collapse
|
36
|
Ricardi U, Badellino S, Filippi AR. What do radiation oncologists require for future advancements in lung SBRT? Phys Med 2016; 44:150-156. [PMID: 27914779 DOI: 10.1016/j.ejmp.2016.11.114] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 11/11/2016] [Accepted: 11/17/2016] [Indexed: 12/25/2022] Open
Abstract
Stereotactic Body Radiotherapy (SBRT) is a well established therapeutic option for patients affected with peripheral early stage non-small cell lung cancer (NSCLC), given the positive clinical evidence accumulated so far on its efficacy and safety. SBRT is regarded as the best choice for inoperable patients, and could also be offered as an alternative to surgery to selected operable patients. More recently, its use for lung metastases progressively increased, and SBRT is now regarded as a low toxic and highly effective local therapy for lung oligometastases from different primary tumors, especially colorectal cancer. Improved planning and delivery techniques have facilitated over the years its use on large and/or centrally located primary tumors, and multiple nodules. Given the successful applications and the current wide dissemination of this technique, clinicians are now faced with an increasingly complex and multi-variable decision process. Some clinically relevant factors are still uncertain, and strategies are needed to reduce the risk of both local and distant failures. Secondly, aspects related to target delineation, dose prescription, image guidance and treatment planning still need to be fully addressed; this may hamper, at least for now, the standardization of SBRT procedures through different Institutions making any kind of direct outcomes comparison difficult. We here aim to provide a perspective on the current role of lung SBRT and its critical aspects, highlighting the potential future developments.
Collapse
|