1
|
Cuccia F, D'Alessandro S, Blasi L, Chiantera V, Ferrera G. The Role of Radiotherapy in the Management of Vaginal Melanoma: A Literature Review with a Focus on the Potential Synergistic Role of Immunotherapy. J Pers Med 2023; 13:1142. [PMID: 37511755 PMCID: PMC10381892 DOI: 10.3390/jpm13071142] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/13/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
Among the mucosal melanomas, vaginal melanomas are very rare tumors, accounting for less than 20% of melanomas arising from the female genital tract. They occur most frequently in women in post-menopausal age, but younger patients may also experience this neoplasm, mainly located in the lower third of the vagina or the anterior wall. The optimal management of this tumor remains controversial, with surgery reported as the most frequently adopted approach. However, a clear benefit of surgical treatment in terms of survival has not yet been demonstrated. Conversely, radiotherapy may represent an attractive non-invasive alternative, and there are several favorable reports of the role of radiation therapy, either delivered with photons, brachytherapy, or hadrontherapy. A wide range of techniques and fractionation regimens are reported with substantially good tolerance to the treatment, and acute G3 or higher toxicities are reported only in the case of concurrent immunotherapy. Of note, due to the rarity of the disease, there is a lack of high-level evidence for the optimal therapeutic option. In this scenario, recent studies theorize the possibility of developing combinatorial approaches of radiotherapy with immunotherapy based on cutaneous melanomas reports. In this review, we aim to summarize the evidence available in the literature supporting the role of definitive radiotherapy for vaginal melanomas, with a focus on the combination of RT with immunotherapy, in terms of optimal timing and biological rationale.
Collapse
Affiliation(s)
- Francesco Cuccia
- Radiotherapy Unit, Radiation Oncology, ARNAS Civico Hospital, 90100 Palermo, Italy
| | - Salvatore D'Alessandro
- Radiotherapy Unit, Radiation Oncology, ARNAS Civico Hospital, 90100 Palermo, Italy
- Radiation Oncology School, University of Palermo, 90100 Palermo, Italy
| | - Livio Blasi
- Medical Oncology, ARNAS Civico Hospital, 90100 Palermo, Italy
| | - Vito Chiantera
- Gynecological Oncology, ARNAS Civico Hospital, 90100 Palermo, Italy
| | - Giuseppe Ferrera
- Radiotherapy Unit, Radiation Oncology, ARNAS Civico Hospital, 90100 Palermo, Italy
| |
Collapse
|
2
|
Dabo-Trubelja A, Gottumukkala V. Review of cancer therapies for the perioperative physician. Perioper Med (Lond) 2023; 12:25. [PMID: 37312150 PMCID: PMC10262136 DOI: 10.1186/s13741-023-00315-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 05/20/2023] [Indexed: 06/15/2023] Open
Abstract
Advances in cancer treatments over the past decades combining chemotherapy with novel technologies in immunotherapies, radiation therapies, and interventional radiology have prolonged life expectancy. Patients have more options for treatments of their primary or metastatic diseases. Increased procedural techniques amid an aging population with multiple comorbidities present risks and challenges in the perioperative period.Chemotherapy remains the mainstay of cancer treatment, can be given intraoperatively, and is combined with other treatment modalities. Immunotherapy is particular to cancer cells while being less toxic to healthy cells. Cancer vaccines stimulate the immune system to stop disease progression. Oncolytic viruses enhance the immune system's cytotoxic effect and show promise to halt metastatic disease progression if present in the perioperative period. Novel techniques in radiation therapy combined with traditional treatments show enhanced survival. This review focuses on current cancer treatments encountered in the perioperative period.
Collapse
Affiliation(s)
- Anahita Dabo-Trubelja
- Department of Anesthesiology and Critical Care, Onco-Anesthesia Fellowship, Perioperative Echocardiography and Ultrasound, Memorial Sloan Kettering Cancer Center of Weill Cornell Medical Center, 1274 York Ave C-330, New York, NY, 10065, USA.
| | - Vijaya Gottumukkala
- Department of Anesthesiology and Perioperative Medicine Program for Advancement of Perioperative Cancer Care, MD Anderson Cancer Center, The University of Texas, Houston, TX, USA
| |
Collapse
|
3
|
Carrasco-Esteban E, Barrionuevo-Castillo P, Domínguez-Rullán J, Gómez-Aparicio MA, Ferri-Molina M, Sáez-Bueno P, Zalabarría-Zarrabeitia Z, Scorsetti M, Arcangeli S, López-Campos F, Couñago F. Stereotactic Body Radiotherapy for Kidney Cancer: Ready for Prime Time? Clin Oncol (R Coll Radiol) 2023; 35:163-176. [PMID: 36443137 DOI: 10.1016/j.clon.2022.11.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 11/09/2022] [Indexed: 11/27/2022]
Abstract
The standard treatment for renal cell carcinoma (RCC) is surgery. However, a number of patients will not be candidates for surgical treatment or will reject this therapeutic approach. Therefore, alternative approaches are required. Historically, radiotherapy has been considered an ineffective treatment for RCC due to the radioresistance of renal tumour cells to conventional fractionation and the increased rate of toxicity. Stereotactic body radiotherapy (SBRT) is a radiotherapy technique that provides a non-invasive ablative treatment with remarkable rates of local control in both primary tumours and metastases in several locations, with a low associated morbidity due to the highly conformal dose and the use of image-guided techniques. Current evidence shows that a higher dose per fraction, achieving a higher biological effective dose, can overcome the radioresistance of RCC cells. Therefore, SBRT, as well as the combination of SBRT and new emerging immune therapies, has a potential role in the local treatment of primary RCC and oligometastatic RCC patients.
Collapse
Affiliation(s)
- E Carrasco-Esteban
- Department of Radiation Oncology, Hospital Central de la Defensa Gómez Ulla, Madrid, Spain
| | | | - J Domínguez-Rullán
- Department of Radiation Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - M A Gómez-Aparicio
- Department of Radiation Oncology, Hospital Universitario de Toledo, Toledo, Spain
| | - M Ferri-Molina
- Department of Radiation Oncology, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - P Sáez-Bueno
- Department of Radiation Oncology, Hospital Central de la Defensa Gómez Ulla, Madrid, Spain
| | | | - M Scorsetti
- Department of Radiotherapy and Radiosurgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - S Arcangeli
- UOC Radioterapia ASST Monza, Università di Milano, Bicocca, Italy
| | - F López-Campos
- Department of Radiation Oncology, Hospital Universitario Ramón y Cajal, Madrid, Spain.
| | - F Couñago
- Department of Radiation Oncology, Hospital Universitario Quirónsalud Madrid, Pozuelo, Madrid, Spain; Universidad Europea de Madrid (UEM), Madrid, Spain
| |
Collapse
|
4
|
Bulgarelli J, Piccinini C, Petracci E, Pancisi E, Granato AM, de Rosa F, Guidoboni M, Petrini M, Ancarani V, Foschi G, Romeo A, Tontini L, De Giorgi U, Lolli C, Gentili G, Valmorri L, Rossi A, Ferroni F, Casadei C, Cortesi P, Crudi L, Ridolfi L. Radiotherapy and High-Dose Interleukin-2: Clinical and Immunological Results of a Proof of Principle Study in Metastatic Melanoma and Renal Cell Carcinoma. Front Immunol 2021; 12:778459. [PMID: 34777395 PMCID: PMC8578837 DOI: 10.3389/fimmu.2021.778459] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 10/13/2021] [Indexed: 11/24/2022] Open
Abstract
High-dose interleukin-2 (HD IL-2) has curative potential in metastatic melanoma (MM) and renal cell carcinoma (RCC). Radiotherapy (RT) kills cancer cells and induces immunomodulatory effects. Prospective trials exploring clinical and immunological properties of combined RT/HD IL-2 are still needed. We designed a phase II, single-arm clinical trial for patients with MM and RCC. The treatment schedule consisted of 3 daily doses of 6-12 Gy of RT to 1-5 non-index metastatic fields, before IL-2 at the first and third treatment cycle. HD IL-2 was administered by continuous infusion for 72 hours and repeated every 3 weeks for up to 4 cycles, thereafter every 4 weeks for a maximum of 2 cycles. The primary endpoint was the immunological efficacy of the combined RT/HD IL-2 treatment (assessed by IFN-γ ELISPOT). Nineteen out of 22 patients were evaluable for immunological and clinical response. Partial response occurred in 3 (15.7%) patients and stable disease was observed in 7 (36.8%). The disease control rate was 52.6% after a median follow up of 39.2 months. According to Common Terminology Criteria for Adverse Events 4.0 (CTCAE 4.0), the majority of toxicities were grade 1-2. Immunological responses were frequent and detected in 16 (84.2%) patients. Increased levels of IL-8 and IL-10 in melanoma, circulating effector memory CD4+ and intratumoral CD8+ T cells in both tumor types were detected after therapy. Overall the treatment was well tolerated and immunologically active. Immunomonitoring and correlative data on tumor and peripheral blood cell subsets suggest that this combination treatment could be a promising strategy for patients progressing after standard treatments.
Collapse
Affiliation(s)
- Jenny Bulgarelli
- Immunotherapy, Cell Therapy and Biobank Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Claudia Piccinini
- Immunotherapy, Cell Therapy and Biobank Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Elisabetta Petracci
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Elena Pancisi
- Immunotherapy, Cell Therapy and Biobank Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Anna Maria Granato
- Immunotherapy, Cell Therapy and Biobank Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Francesco de Rosa
- Immunotherapy, Cell Therapy and Biobank Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Massimo Guidoboni
- Immunotherapy, Cell Therapy and Biobank Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Massimiliano Petrini
- Immunotherapy, Cell Therapy and Biobank Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Valentina Ancarani
- Immunotherapy, Cell Therapy and Biobank Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giovanni Foschi
- Immunotherapy, Cell Therapy and Biobank Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Antonino Romeo
- Radiotherapy Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Luca Tontini
- Radiotherapy Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Ugo De Giorgi
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Cristian Lolli
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Giorgia Gentili
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Linda Valmorri
- Unit of Biostatistics and Clinical Trials, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Alice Rossi
- Radiology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Fabio Ferroni
- Radiology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Carla Casadei
- Anesthesiology Service, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Pietro Cortesi
- Cardio-Oncology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Laura Crudi
- Oncology Pharmacy Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Laura Ridolfi
- Immunotherapy, Cell Therapy and Biobank Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
5
|
Sezen D, Patel RR, Tang C, Onstad M, Nagarajan P, Patel SP, Welsh JW, Lin LL. Immunotherapy combined with high- and low-dose radiation to all sites leads to complete clearance of disease in a patient with metastatic vaginal melanoma. Gynecol Oncol 2021; 161:645-652. [PMID: 33795130 DOI: 10.1016/j.ygyno.2021.03.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
A 73-year-old woman with metastatic vaginal mucosal melanoma that had progressed on ipilimumab and nivolumab experienced clinical and radiographic complete response to dual checkpoint inhibitor immunotherapy given in combination with high-dose plus low-dose radiation. General characteristics and treatment options in this disease are highlighted.
Collapse
Affiliation(s)
- Duygu Sezen
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Radiation Oncology, Koc University School of Medicine, Istanbul, Turkey
| | - Roshal R Patel
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Albany Medical College, Albany, NY, USA
| | - Christine Tang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Baylor College of Medicine, Houston, TX, USA
| | - Michaela Onstad
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Priyadharsini Nagarajan
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sapna P Patel
- Department of Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - James W Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lilie L Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
6
|
Li Q, Li J, Song S, Chen W, Shen X, Li S, Xing D. Nanoparticle-mediated tumor vaccines for personalized therapy: preparing tumor antigens in vivo or ex vivo? J Mater Chem B 2021; 9:2352-2366. [PMID: 33659970 DOI: 10.1039/d0tb02915g] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Tumor vaccines, focusing on tailoring individual tumor antigens, have gained much attention in personalized tumor therapy. Recently, breakthroughs have been made in the development of tumor vaccines thanks to the progress in nanotechnology. We will summarize nanoparticle-mediated tumor vaccines for personalized therapy in this review. ROS/heat generating nanoparticles and molecules could induce immunogenic cell death and tumor antigen release in vivo. This strategy often includes chemotherapy, radiotherapy, photodynamic therapy, photothermal therapy, magneto-thermal therapy, etc. On the other hand, ex vivo technologies have been applied for processing of tumor cells/tissues to form effective tumor antigens, in which nanotechnology has shown very good prospects in delivering tumor antigens. In in vivo and ex vivo strategies, nanotechnology also could improve the immune effect through enhancing the uptake by targeting cells, reducing therapeutic drugs/agents, further encapsulating immuno-modulatory molecules or combining with other therapy treatments. Thus, therapeutic vaccines based on nanoparticles have the potential to enhance the immune response and reduce the side effects.
Collapse
Affiliation(s)
- Qian Li
- Cancer Institute, Affiliated Hospital of Qingdao University, Qingdao, 266071, China and Qingdao Cancer Institute, Qingdao, 266071, China
| | - Jia Li
- Qingdao Cancer Institute, Qingdao, 266071, China
| | - Sha Song
- Qingdao Cancer Institute, Qingdao, 266071, China
| | - Wujun Chen
- Qingdao Cancer Institute, Qingdao, 266071, China
| | - Xin Shen
- Cancer Institute, Affiliated Hospital of Qingdao University, Qingdao, 266071, China and CP Pharmaceutical (Qingdao) Co., Ltd, Qingdao, 266426, China.
| | - Suming Li
- Institut Européen des Membranes, IEM-UMR 5635, Univ Montpellier, ENSCM,CNRS, 34095 Montpellier, France.
| | - Dongming Xing
- Cancer Institute, Affiliated Hospital of Qingdao University, Qingdao, 266071, China and Qingdao Cancer Institute, Qingdao, 266071, China and School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
7
|
Antibody-Based Immunotherapy: Alternative Approaches for the Treatment of Metastatic Melanoma. Biomedicines 2020; 8:biomedicines8090327. [PMID: 32899183 PMCID: PMC7555584 DOI: 10.3390/biomedicines8090327] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/28/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022] Open
Abstract
Melanoma is the least common form of skin cancer and is associated with the highest mortality. Where melanoma is mostly unresponsive to conventional therapies (e.g., chemotherapy), BRAF inhibitor treatment has shown improved therapeutic outcomes. Photodynamic therapy (PDT) relies on a light-activated compound to produce death-inducing amounts of reactive oxygen species (ROS). Their capacity to selectively accumulate in tumor cells has been confirmed in melanoma treatment with some encouraging results. However, this treatment approach has not reached clinical fruition for melanoma due to major limitations associated with the development of resistance and subsequent side effects. These adverse effects might be bypassed by immunotherapy in the form of antibody–drug conjugates (ADCs) relying on the ability of monoclonal antibodies (mAbs) to target specific tumor-associated antigens (TAAs) and to be used as carriers to specifically deliver cytotoxic warheads into corresponding tumor cells. Of late, the continued refinement of ADC therapeutic efficacy has given rise to photoimmunotherapy (PIT) (a light-sensitive compound conjugated to mAbs), which by virtue of requiring light activation only exerts its toxic effect on light-irradiated cells. As such, this review aims to highlight the potential clinical benefits of various armed antibody-based immunotherapies, including PDT, as alternative approaches for the treatment of metastatic melanoma.
Collapse
|
8
|
Wang J, Li Z, Wang Z, Yu Y, Li D, Li B, Ding J. Nanomaterials for Combinational Radio–Immuno Oncotherapy. ADVANCED FUNCTIONAL MATERIALS 2020; 30. [DOI: 10.1002/adfm.201910676] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 03/09/2020] [Indexed: 08/29/2023]
Abstract
AbstractRadiotherapy, a clinically used local treatment modality of cancers, is regarded as a promising candidate to promote current immunotherapy through initiating an in situ vaccination effect and reprogramming the immunosuppressive microenvironment. The combination of radiotherapy and immunotherapy, referred to as combinational radio–immuno oncotherapy (CRIOT), elicits a synergistic antitumor effect based on the immunomodulatory properties of radiation. Unfortunately, current CRIOT accompanies low response rate and severe toxicity in clinical trials, thus limiting its application. To this end, various nanomaterials are being developed to sensitize radiotherapy or deliver immune agents, or both, to improve the unsatisfactory outcomes of CRIOT. Herein, enhanced antitumor efficacy of CRIOT with nanomaterials through the possible mechanisms of rejuvenation and activation of T cells, increased presentation of tumor‐related antigens, and inhibition of suppressive macrophages is presented, and the prospect of CRIOT in clinical practice is proposed.
Collapse
Affiliation(s)
- Juan Wang
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
- Department of Radiation Oncology Cancer Hospital of Shandong First Medical University 440 Jiyan Road Jinan 250117 P. R. China
| | - Zhongmin Li
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
- Department of Gastrointestinal, Colorectal, and Anal Surgery China–Japan Union Hospital of Jilin University 126 Xiantai Street Changchun 130012 P. R. China
| | - Zhongtang Wang
- Department of Radiation Oncology Cancer Hospital of Shandong First Medical University 440 Jiyan Road Jinan 250117 P. R. China
| | - Yonghua Yu
- Department of Radiation Oncology Cancer Hospital of Shandong First Medical University 440 Jiyan Road Jinan 250117 P. R. China
| | - Di Li
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
| | - Baosheng Li
- Department of Radiation Oncology Cancer Hospital of Shandong First Medical University 440 Jiyan Road Jinan 250117 P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials Changchun Institute of Applied Chemistry Chinese Academy of Sciences 5625 Renmin Street Changchun 130022 P. R. China
| |
Collapse
|
9
|
Chulpanova DS, Kitaeva KV, Green AR, Rizvanov AA, Solovyeva VV. Molecular Aspects and Future Perspectives of Cytokine-Based Anti-cancer Immunotherapy. Front Cell Dev Biol 2020; 8:402. [PMID: 32582698 PMCID: PMC7283917 DOI: 10.3389/fcell.2020.00402] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 05/01/2020] [Indexed: 12/11/2022] Open
Abstract
Cytokine-based immunotherapy is a promising field in the cancer treatment, since cytokines, as proteins of the immune system, are able to modulate the host immune response toward cancer cell, as well as directly induce tumor cell death. Since a low dose monotherapy with some cytokines has no significant therapeutic results and a high dose treatment leads to a number of side effects caused by the pleiotropic effect of cytokines, the problem of understanding the influence of cytokines on the immune cells involved in the pro- and anti-tumor immune response remains a pressing one. Immune system cells carry CD makers on their surface which can be used to identify various populations of cells of the immune system that play different roles in pro- and anti-tumor immune responses. This review discusses the functions and specific CD markers of various immune cell populations which are reported to participate in the regulation of the immune response against the tumor. The results of research studies and clinical trials investigating the effect of cytokine therapy on the regulation of immune cell populations and their surface markers are also discussed. Current trends in the development of cancer immunotherapy, as well as the role of cytokines in combination with other therapeutic agents, are also discussed.
Collapse
Affiliation(s)
- Daria S Chulpanova
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Kristina V Kitaeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Andrew R Green
- Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, United Kingdom
| | - Albert A Rizvanov
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Nottingham Breast Cancer Research Centre, Division of Cancer and Stem Cells, School of Medicine, University of Nottingham Biodiscovery Institute, Nottingham, United Kingdom
| | - Valeriya V Solovyeva
- Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
10
|
Palata O, Hradilova Podzimkova N, Nedvedova E, Umprecht A, Sadilkova L, Palova Jelinkova L, Spisek R, Adkins I. Radiotherapy in Combination With Cytokine Treatment. Front Oncol 2019; 9:367. [PMID: 31179236 PMCID: PMC6538686 DOI: 10.3389/fonc.2019.00367] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 04/23/2019] [Indexed: 12/22/2022] Open
Abstract
Radiotherapy (RT) plays an important role in the management of cancer patients. RT is used in more than 50% of patients during the course of their disease in a curative or palliative setting. In the past decades it became apparent that the abscopal effect induced by RT might be dependent on the activation of immune system, and that the induction of immunogenic cancer cell death and production of danger-associated molecular patterns from dying cells play a major role in the radiotherapy-mediated anti-tumor efficacy. Therefore, the combination of RT and immunotherapy is of a particular interest that is reflected in designing clinical trials to treat patients with various malignancies. The use of cytokines as immunoadjuvants in combination with RT has been explored over the last decades as one of the immunotherapeutic combinations to enhance the clinical response to anti-cancer treatment. Here we review mainly the data on the efficacy of IFN-α, IL-2, IL-2-based immunocytokines, GM-CSF, and TNF-α used in combinations with various radiotherapeutic techniques in clinical trials. Moreover, we discuss the potential of IL-15 and its analogs and IL-12 cytokines in combination with RT based on the efficacy in preclinical mouse tumor models.
Collapse
Affiliation(s)
- Ondrej Palata
- SOTIO a.s, Prague, Czechia.,Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czechia
| | - Nada Hradilova Podzimkova
- SOTIO a.s, Prague, Czechia.,Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czechia
| | | | | | | | - Lenka Palova Jelinkova
- SOTIO a.s, Prague, Czechia.,Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czechia
| | - Radek Spisek
- SOTIO a.s, Prague, Czechia.,Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czechia
| | - Irena Adkins
- SOTIO a.s, Prague, Czechia.,Department of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czechia
| |
Collapse
|
11
|
Leaman Alcibar O, Candini D, López-Campos F, Albert Antequera M, Morillo Macías V, Conde AJ, Rodríguez Pérez A, Hervás Morón A, Contreras Martínez J, Ferrer Albiach C, Navarro Aguilar S, Rodríguez-Ruiz ME. Time for radioimmunotherapy: an overview to bring improvements in clinical practice. Clin Transl Oncol 2019; 21:992-1004. [PMID: 30644044 DOI: 10.1007/s12094-018-02027-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 12/21/2018] [Indexed: 12/27/2022]
Abstract
Harnessing the patient's own immune system against an established cancer has proven to be a successful strategy. Within the last years, several antibodies blocking critical "checkpoints" that control the activation of T cells, the immune cells able to kill cancer cells, have been approved for the use in patients with different tumours. Unfortunately, these cases remain a minority. Over the last years, radiotherapy has been reported as a means to turn a patient's own tumour into an in situ vaccine and generate anti-tumour T cells in patients who lack sufficient anti-tumour immunity. Indeed, review data show that the strategy of blocking multiple selected immune inhibitory targets in combination with radiotherapy has the potential to unleash powerful anti-tumour responses and improve the outcome of metastatic solid tumours. Here, we review the principal tumours where research in this field has led to new knowledge and where radioimmunotherapy becomes a reality.
Collapse
Affiliation(s)
- O Leaman Alcibar
- Radiation Oncology Department, Central University Hospital of Defence Gómez Ulla, Madrid, Spain.
| | - D Candini
- Radiation Oncology Department, Ramón y Cajal University Hospital, Madrid, Spain
| | - F López-Campos
- Radiation Oncology Department, Ramón y Cajal University Hospital, Madrid, Spain
| | - M Albert Antequera
- Radiation Oncology Department, La Ribera University Hospital, Valencia, Spain
| | - V Morillo Macías
- Radiation Oncology Department, General Hospital in Castellón de la Plana, Castellón, Spain
| | - A J Conde
- Radiation Oncology Department, The University and Polytechnic La Fe Hospital, Valencia, Spain
| | - A Rodríguez Pérez
- Radiation Oncology Department, Internacional Ruber Hospital, Madrid, Spain
| | - A Hervás Morón
- Radiation Oncology Department, Ramón y Cajal University Hospital, Madrid, Spain
| | | | - C Ferrer Albiach
- Radiation Oncology Department, General Hospital in Castellón de la Plana, Castellón, Spain
| | - S Navarro Aguilar
- Radiation Oncology Department, Oncology and Radiotherapy Institut, Ricardo Palma Clinic, Lima, Peru
| | - M E Rodríguez-Ruiz
- Radiation Oncology Department, University of Navarra Clinic and CIMA, Navarra, Spain
| |
Collapse
|
12
|
Goedegebuure RSA, de Klerk LK, Bass AJ, Derks S, Thijssen VLJL. Combining Radiotherapy With Anti-angiogenic Therapy and Immunotherapy; A Therapeutic Triad for Cancer? Front Immunol 2019; 9:3107. [PMID: 30692993 PMCID: PMC6339950 DOI: 10.3389/fimmu.2018.03107] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/17/2018] [Indexed: 12/19/2022] Open
Abstract
Radiotherapy has been used for the treatment of cancer for over a century. Throughout this period, the therapeutic benefit of radiotherapy has continuously progressed due to technical developments and increased insight in the biological mechanisms underlying the cellular responses to irradiation. In order to further improve radiotherapy efficacy, there is a mounting interest in combining radiotherapy with other forms of therapy such as anti-angiogenic therapy or immunotherapy. These strategies provide different opportunities and challenges, especially with regard to dose scheduling and timing. Addressing these issues requires insight in the interaction between the different treatment modalities. In the current review, we describe the basic principles of the effects of radiotherapy on tumor vascularization and tumor immunity and vice versa. We discuss the main strategies to combine these treatment modalities and the hurdles that have to be overcome in order to maximize therapeutic effectivity. Finally, we evaluate the outstanding questions and present future prospects of a therapeutic triad for cancer.
Collapse
Affiliation(s)
- Ruben S A Goedegebuure
- Amsterdam UMC, Location VUmc, Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Leonie K de Klerk
- Amsterdam UMC, Location VUmc, Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands.,Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Adam J Bass
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States.,Cancer Program, The Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Sarah Derks
- Amsterdam UMC, Location VUmc, Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Victor L J L Thijssen
- Amsterdam UMC, Location VUmc, Medical Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands.,Amsterdam UMC, Location VUmc, Radiation Oncology, Cancer Center Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
13
|
Abstract
The relatively high DNA mutational burden in melanoma allows for the creation of potentially "foreign," immune-stimulating neoantigens, and leads to its exceptional immunogenicity. Brisk tumor-infiltrating lymphocytes, a marker of immune editing, confer improved overall survival in melanoma, possibly due to reduced sentinel lymph node spread. Meanwhile, T-cell-stimulating drugs, so-called T-cell checkpoint inhibitors, which reverse peripheral tolerance-dependent tumor escape, have demonstrated unparalleled clinical success in metastatic melanoma. Markers to predict response to immunotherapy are currently imperfect, and the subject of intense research, which will guide the future of ancillary pathologic testing in this setting.
Collapse
Affiliation(s)
- Jennifer S Ko
- Department of Anatomic Pathology, Cleveland Clinic, 9500 Euclid Avenue, L2-150, Cleveland, OH 44195, USA.
| |
Collapse
|
14
|
Farber SH, Elsamadicy AA, Atik AF, Suryadevara CM, Chongsathidkiet P, Fecci PE, Sampson JH. The Safety of available immunotherapy for the treatment of glioblastoma. Expert Opin Drug Saf 2017; 16:277-287. [PMID: 27989218 DOI: 10.1080/14740338.2017.1273898] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION Glioblastoma (GBM) is the most common malignant primary brain tumor in adults. Current standard of care involves maximal surgical resection combined with adjuvant chemoradiation. Growing support exists for a role of immunotherapy in treating these tumors with the goal of targeted cytotoxicity. Here we review data on the safety for current immunotherapies being tested in GBM. Areas covered: Safety data from published clinical trials, including ongoing clinical trials were reviewed. Immunotherapeutic classes currently under investigation in GBM include various vaccination strategies, adoptive T cell immunotherapy, immune checkpoint blockade, monoclonal antibodies, and cytokine therapies. Trials include children, adolescents, and adults with either primary or recurrent GBM. Expert opinion: Based on the reviewed clinical trials, the current immunotherapies targeting GBM are safe and well-tolerated with minimal toxicities which should be noted. However, the gains in patient survival have been modest. A safe and well-tolerated combinatory immunotherapeutic approach may be essential for optimal efficacy towards GBM.
Collapse
Affiliation(s)
- S Harrison Farber
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b The Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , NC , USA
| | - Aladine A Elsamadicy
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b The Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , NC , USA
| | - Ahmet Fatih Atik
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b The Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , NC , USA
| | - Carter M Suryadevara
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b The Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , NC , USA.,c Department of Pathology , Duke University Medical Center , Durham , NC , USA
| | - Pakawat Chongsathidkiet
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b The Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , NC , USA.,c Department of Pathology , Duke University Medical Center , Durham , NC , USA
| | - Peter E Fecci
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b The Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , NC , USA.,c Department of Pathology , Duke University Medical Center , Durham , NC , USA
| | - John H Sampson
- a Duke Brain Tumor Immunotherapy Program, Department of Neurosurgery , Duke University Medical Center , Durham , NC , USA.,b The Preston Robert Tisch Brain Tumor Center , Duke University Medical Center , Durham , NC , USA.,c Department of Pathology , Duke University Medical Center , Durham , NC , USA
| |
Collapse
|
15
|
Schoenhals JE, Skrepnik T, Selek U, Cortez MA, Li A, Welsh JW. Optimizing Radiotherapy with Immunotherapeutic Approaches. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 995:53-71. [PMID: 28321812 DOI: 10.1007/978-3-319-53156-4_3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Several factors must be considered to successfully integrate immunotherapy with radiation into clinical practice. One such factor is that concepts arising from preclinical work must be tested in combination with radiation in preclinical models to better understand how combination therapy will work in patients; examples include checkpoint inhibitors, tumor growth factor-beta (TGF-β) inhibitors, and natural killer (NK) cell therapy. Also, many radiation fields and fractionation schedules typically used in radiation therapy had been standardized before the introduction of advanced techniques for radiation planning and delivery that account for changes in tumor size, location, and motion during treatment, as well as uncertainties introduced by variations in patient setup between treatment fractions. As a result, radiation therapy may involve the use of large treatment volumes, often encompassing nodal regions that may not be irradiated with more conformal techniques. Traditional forms of radiation in particular pose challenges for combination trials with immunotherapy. This chapter explores these issues in more detail and provides insights as to how radiation therapy can be optimized to combine with immunotherapy.
Collapse
Affiliation(s)
- Jonathan E Schoenhals
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tijana Skrepnik
- Department of Radiation Oncology, University of Arizona, Tucson, AZ, USA
| | - Ugur Selek
- Department of Radiation Oncology, Koc University, Istanbul, Turkey
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Anderson Central (Y2.5316), 1515 Holcombe Blvd., Unit 0097, Houston, TX, 77030, USA
| | - Maria A Cortez
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ailin Li
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China
| | - James W Welsh
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Anderson Central (Y2.5316), 1515 Holcombe Blvd., Unit 0097, Houston, TX, 77030, USA.
| |
Collapse
|
16
|
Malignant melanoma—The cradle of anti-neoplastic immunotherapy. Crit Rev Oncol Hematol 2016; 106:25-54. [DOI: 10.1016/j.critrevonc.2016.04.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 03/14/2016] [Accepted: 04/25/2016] [Indexed: 02/07/2023] Open
|
17
|
Wargo JA, Reuben A, Cooper ZA, Oh KS, Sullivan RJ. Immune Effects of Chemotherapy, Radiation, and Targeted Therapy and Opportunities for Combination With Immunotherapy. Semin Oncol 2015; 42:601-16. [PMID: 26320064 DOI: 10.1053/j.seminoncol.2015.05.007] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
There have been significant advances in cancer treatment over the past several years through the use of chemotherapy, radiation therapy, molecularly targeted therapy, and immunotherapy. Despite these advances, treatments such as monotherapy or monomodality have significant limitations. There is increasing interest in using these strategies in combination; however, it is not completely clear how best to incorporate molecularly targeted and immune-targeted therapies into combination regimens. This is particularly pertinent when considering combinations with immunotherapy, as other types of therapy may have significant impact on host immunity, the tumor microenvironment, or both. Thus, the influence of chemotherapy, radiation therapy, and molecularly targeted therapy on the host anti-tumor immune response and the host anti-host response (ie, autoimmune toxicity) must be taken into consideration when designing immunotherapy-based combination regimens. We present data related to many of these combination approaches in the context of investigations in patients with melanoma and discuss their potential relationship to management of patients with other tumor types. Importantly, we also highlight challenges of these approaches and emphasize the need for continued translational research.
Collapse
Affiliation(s)
- Jennifer A Wargo
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX; Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Alexandre Reuben
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Zachary A Cooper
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX; Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kevin S Oh
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Ryan J Sullivan
- Department of Medical Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA.
| |
Collapse
|
18
|
Hecht M, Zimmer L, Loquai C, Weishaupt C, Gutzmer R, Schuster B, Gleisner S, Schulze B, Goldinger SM, Berking C, Forschner A, Clemens P, Grabenbauer G, Müller-Brenne T, Bauch J, Eich HT, Grabbe S, Schadendorf D, Schuler G, Keikavoussi P, Semrau S, Fietkau R, Distel LV, Heinzerling L. Radiosensitization by BRAF inhibitor therapy-mechanism and frequency of toxicity in melanoma patients. Ann Oncol 2015; 26:1238-1244. [PMID: 25762352 DOI: 10.1093/annonc/mdv139] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2015] [Accepted: 03/02/2015] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND Recent evidence suggests that ionizing radiation may be associated with unexpected side-effects in melanoma patients treated with concomitant BRAF inhibitors. A large multicenter analysis was carried out to generate reliable safety data and elucidate the mechanism. METHODS A total of 161 melanoma patients from 11 European skin cancer centers were evaluated for acute and late toxicity, of whom 70 consecutive patients received 86 series of radiotherapy with concomitant BRAF inhibitor therapy. To further characterize and quantify a possible radiosensitization by BRAF inhibitors, blood samples of 35 melanoma patients were used for individual radiosensitivity testing by fluorescence in situ hybridization of chromosomal breaks after ex vivo irradiation. RESULTS With radiotherapy and concomitant BRAF inhibitor therapy the rate of acute radiodermatitis ≥2° was 36% and follicular cystic proliferation was seen in 13% of all radiotherapies. Non-skin toxicities included hearing disorders (4%) and dysphagia (2%). Following whole-brain radiotherapy, rates of radiodermatitis ≥2° were 44% and 8% (P < 0.001) for patients with and without BRAF inhibitor therapy, respectively. Concomitant treatment with vemurafenib induced acute radiodermatitis ≥2° more frequently than treatment with dabrafenib (40% versus 26%, P = 0.07). In line with these findings, analysis of chromosomal breaks ex vivo indicated significantly increased radiosensitivity for patients under vemurafenib (P = 0.004) and for patients switched from vemurafenib to dabrafenib (P = 0.002), but not for patients on dabrafenib only. No toxicities were reported after stereotactic treatment. CONCLUSION Radiotherapy with concomitant BRAF inhibitor therapy is feasible with an acceptable increase in toxicity. Vemurafenib is a more potent radiosensitizer than dabrafenib.
Collapse
Affiliation(s)
- M Hecht
- Department of Radiation Oncology, University Hospital Erlangen, Erlangen
| | - L Zimmer
- Department of Dermatology, University Hospital Essen, Essen
| | - C Loquai
- Department of Dermatology, University Hospital Mainz, Mainz
| | - C Weishaupt
- Department of Dermatology, University Hospital Münster, Münster
| | - R Gutzmer
- Department of Dermatology, Hannover Medical School, Hannover
| | - B Schuster
- Department of Radiation Oncology, University Hospital Erlangen, Erlangen
| | - S Gleisner
- Department of Radiation Oncology, University Hospital Erlangen, Erlangen
| | - B Schulze
- Department of Radiation Oncology, University Hospital Frankfurt, Frankfurt, Germany
| | - S M Goldinger
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - C Berking
- Department of Dermatology, University Hospital München (LMU), München
| | - A Forschner
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - P Clemens
- Department of Radiation Oncology, Hospital Feldkirch, Feldkirch, Austria
| | - G Grabenbauer
- Department of Radiation Oncology, Hospital Coburg, Coburg
| | - T Müller-Brenne
- Department of Radiation Oncology, University Hospital Mainz, Mainz
| | - J Bauch
- Department of Radiation Oncology, University Hospital Münster, Münster
| | - H T Eich
- Department of Radiation Oncology, University Hospital Münster, Münster
| | - S Grabbe
- Department of Dermatology, University Hospital Mainz, Mainz
| | - D Schadendorf
- Department of Dermatology, University Hospital Essen, Essen
| | - G Schuler
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - P Keikavoussi
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| | - S Semrau
- Department of Radiation Oncology, University Hospital Erlangen, Erlangen
| | - R Fietkau
- Department of Radiation Oncology, University Hospital Erlangen, Erlangen
| | - L V Distel
- Department of Radiation Oncology, University Hospital Erlangen, Erlangen.
| | - L Heinzerling
- Department of Dermatology, University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|