4151
|
The Rapid Emergence of Novel Therapeutics in Advanced Malignant Melanoma. Dermatol Ther (Heidelb) 2015; 5:151-69. [PMID: 26387031 PMCID: PMC4580658 DOI: 10.1007/s13555-015-0080-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Indexed: 02/07/2023] Open
Abstract
For decades, no cancer therapy had been shown to improve average survival in metastatic melanoma. Two critical events have occurred, the discovery of melanoma driver mutation subsets and the discovery of immune checkpoint inhibitors, which have allowed for the development of modern, effective therapies. These findings have facilitated a rapid emergence of novel therapeutics for the disease with multiple FDA approvals in the last several years. The drugs vemurafenib, trametinib, and dabrafenib, which inhibit the commonly mutated BRAF pathway, have been approved based on improvements in survival outcomes. Agents that block immune checkpoints on lymphocytes allowing for immune cell activity against melanoma have also been approved based on improved survival outcomes such as ipilimumab and nivolumab. Pembrolizumab, another immune checkpoint inhibitor, has also been approved based on the response rate and duration of response in a phase 1 trial. Further agents and combinations of approved agents are positioned to possibly further increase this tally of approved drugs. This review will discuss recently approved novel agents and select drugs in development in advanced melanoma.
Collapse
|
4152
|
Watanabe M, Yamamoto H, Hashida S, Soh J, Sugimoto S, Toyooka S, Miyoshi S. Primary pulmonary melanoma: a report of two cases. World J Surg Oncol 2015; 13:274. [PMID: 26376781 PMCID: PMC4573480 DOI: 10.1186/s12957-015-0695-2] [Citation(s) in RCA: 9] [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: 04/27/2015] [Accepted: 09/07/2015] [Indexed: 01/21/2023] Open
Abstract
Malignant melanoma is a refractory malignancy with a dismal prognosis. It generally arises from the skin in most cases, and cases of primary pulmonary malignant melanoma are rare and often behave aggressively. We have treated two cases of localized primary pulmonary malignant melanoma using surgical resection. Pulmonary malignant melanomas often metastasize to the brain and liver; one of our cases exhibited metastasis to the cecum at about 8 months after surgery. Because cutaneous melanomas often carry activating mutations in the BRAF gene (V600E), we performed a BRAF mutational analysis using direct sequencing for both of these tumors arising from the lung. However, no BRAF mutations were detected. We detected a p53 mutation, which was thought to be a potential somatic mutation, in one of the two cases using a sequencing panel targeting 20 lung cancer-related genes. Although we also checked the expression of programmed death ligand 1 (PD-L1) on the surface of the tumor cells by immunohistochemical testing, neither of our two cases expressed PD-L1. Further molecular analyses may uncover the characteristics of primary pulmonary malignant melanomas.
Collapse
Affiliation(s)
- Mototsugu Watanabe
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan.
| | - Hiromasa Yamamoto
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Shinsuke Hashida
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
- Department of Clinical Genomic Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan
| | - Junichi Soh
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
- Biobank of Okayama University Hospital, Okayama, 700-8558, Japan
| | - Seiichiro Sugimoto
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Shinichi Toyooka
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan.
- Department of Clinical Genomic Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama, 700-8558, Japan.
- Biobank of Okayama University Hospital, Okayama, 700-8558, Japan.
| | - Shinichiro Miyoshi
- Department of Thoracic, Breast and Endocrinological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| |
Collapse
|
4153
|
Heterocyclic Anticancer Compounds: Recent Advances and the Paradigm Shift towards the Use of Nanomedicine's Tool Box. Molecules 2015; 20:16852-91. [PMID: 26389876 PMCID: PMC6331900 DOI: 10.3390/molecules200916852] [Citation(s) in RCA: 385] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 09/08/2015] [Accepted: 09/09/2015] [Indexed: 02/07/2023] Open
Abstract
The majority of heterocycle compounds and typically common heterocycle fragments present in most pharmaceuticals currently marketed, alongside with their intrinsic versatility and unique physicochemical properties, have poised them as true cornerstones of medicinal chemistry. Apart from the already marketed drugs, there are many other being investigated for their promising activity against several malignancies. In particular, anticancer research has been capitalizing on the intrinsic versatility and dynamic core scaffold of these compounds. Nevertheless, as for any other promising anticancer drugs, heterocyclic compounds do not come without shortcomings. In this review, we provide for a concise overview of heterocyclic active compounds and families and their main applications in medicine. We shall focus on those suitable for cancer therapy while simultaneously addressing main biochemical modes of action, biological targets, structure-activity relationships as well as intrinsic limitation issues in the use of these compounds. Finally, considering the advent of nanotechnology for effective selective targeting of drugs, we shall discuss fundamental aspects and considerations on nanovectorization of such compounds that may improve pharmacokinetic/pharmacodynamic properties of heterocycles.
Collapse
|
4154
|
Coffelt SB, de Visser KE. Immune-mediated mechanisms influencing the efficacy of anticancer therapies. Trends Immunol 2015; 36:198-216. [PMID: 25857662 DOI: 10.1016/j.it.2015.02.006] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 02/18/2015] [Accepted: 02/18/2015] [Indexed: 01/26/2023]
Abstract
Conventional anticancer therapies, such as chemotherapy, radiotherapy, and targeted therapy, are designed to kill cancer cells. However, the efficacy of anticancer therapies is not only determined by their direct effects on cancer cells but also by off-target effects within the host immune system. Cytotoxic treatment regimens elicit several changes in immune-related parameters including the composition, phenotype, and function of immune cells. Here we discuss the impact of innate and adaptive immune cells on the success of anticancer therapy. In this context we examine the opportunities to exploit host immune responses to boost tumor clearing, and highlight the challenges facing the treatment of advanced metastatic disease.
Collapse
Affiliation(s)
- Seth B Coffelt
- Division of Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| | - Karin E de Visser
- Division of Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| |
Collapse
|
4155
|
Imafuku K, Yoshino K, Ishiwata K, Otobe S, Tsuboi S, Ohara K, Hata H. Severe rash associated with vemurafenib administration following nivolumab therapy. J Eur Acad Dermatol Venereol 2015; 30:e84-e86. [PMID: 26372833 DOI: 10.1111/jdv.13331] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- K Imafuku
- Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan.
| | - K Yoshino
- Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - K Ishiwata
- Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - S Otobe
- Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - S Tsuboi
- Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - K Ohara
- Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - H Hata
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
4156
|
Carlino MS, Long GV. Is chemotherapy still an option in the treatment of melanoma? Ann Oncol 2015; 26:2203-4. [PMID: 26374287 DOI: 10.1093/annonc/mdv361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- M S Carlino
- Melanoma Institute Australia, Sydney The Sydney Medical School, The University of Sydney, Sydney Department of Medical Oncology, Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney
| | - G V Long
- Melanoma Institute Australia, Sydney The Sydney Medical School, The University of Sydney, Sydney The Mater Hospital, North Sydney, Australia
| |
Collapse
|
4157
|
Sudo K, Yamada Y. Advancing pharmacological treatment options for advanced gastric cancer. Expert Opin Pharmacother 2015; 16:2293-305. [PMID: 26359224 DOI: 10.1517/14656566.2015.1080238] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
INTRODUCTION Gastric cancer is the third most common cause of cancer-related deaths worldwide. Improvement of conventional chemotherapy has been modest in the past decades. AREAS COVERED We review recent important studies of metastatic or recurrent gastric cancer. For human epidermal growth factor receptors 2 (HER2) negative cancer, standard treatments are combinations of fluoropyrimidine and platinum with or without epirubicin or docetaxel in first-line therapy. Controversy exists regarding the use of triplet chemotherapies due to their toxicity. For HER2 positive cancer, standard treatments are combinations of fluoropyrimidine and cisplatin with trastuzumab. As second- or third-line treatment, taxanes or irinotecan prolonged survival compared with best supportive care alone, but the extension of overall survival was only 1 - 2 months. A recent study demonstrated that ramucirumab plus paclitaxel improved survival as a second-line therapy. EXPERT OPINION Most trials have failed to demonstrate a benefit of targeted agents. It is important to identify predictive biomarkers to enrich an appropriate patient population for targeted agents such as HER2 status for trastuzumab.
Collapse
Affiliation(s)
- Kazuki Sudo
- a 1 National Cancer Center Hospital , 5-1-1 Tsukiji, Chuo-ku, 104-0045 Tokyo, Japan +81 3 3542 2511 ; +81 3 3542 3815 ; .,b 2 Juntendo University Graduate School of Medicine, Advanced Clinical Research of Cancer , Tokyo, Japan
| | - Yasuhide Yamada
- a 1 National Cancer Center Hospital , 5-1-1 Tsukiji, Chuo-ku, 104-0045 Tokyo, Japan +81 3 3542 2511 ; +81 3 3542 3815 ;
| |
Collapse
|
4158
|
Smith HG, Cartwright J, Wilkinson MJ, Strauss DC, Thomas JM, Hayes AJ. Isolated Limb Perfusion with Melphalan and Tumour Necrosis Factor α for In-Transit Melanoma and Soft Tissue Sarcoma. Ann Surg Oncol 2015; 22 Suppl 3:S356-61. [DOI: 10.1245/s10434-015-4856-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Indexed: 11/18/2022]
|
4159
|
Appleton ES, Turnbull S, Ralph C, West E, Scott K, Harrington K, Pandha H, Melcher A. Talimogene laherparepvec in the treatment of melanoma. Expert Opin Biol Ther 2015; 15:1517-30. [DOI: 10.1517/14712598.2015.1084280] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
4160
|
Dissecting the Potential Interplay of DEK Functions in Inflammation and Cancer. JOURNAL OF ONCOLOGY 2015; 2015:106517. [PMID: 26425120 PMCID: PMC4575739 DOI: 10.1155/2015/106517] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 03/05/2015] [Indexed: 12/12/2022]
Abstract
There is a long-standing correlation between inflammation, inflammatory cell signaling pathways, and tumor formation. Understanding the mechanisms behind inflammation-driven tumorigenesis is of great research and clinical importance. Although not entirely understood, these mechanisms include a complex interaction between the immune system and the damaged epithelium that is mediated by an array of molecular signals of inflammation—including reactive oxygen species (ROS), cytokines, and NFκB signaling—that are also oncogenic. Here, we discuss the association of the unique DEK protein with these processes. Specifically, we address the role of DEK in chronic inflammation via viral infections and autoimmune diseases, the overexpression and oncogenic activity of DEK in cancers, and DEK-mediated regulation of NFκB signaling. Combined, evidence suggests that DEK may play a complex, multidimensional role in chronic inflammation and subsequent tumorigenesis.
Collapse
|
4161
|
Wu J, Fu J, Zhang M, Liu D. Blinatumomab: a bispecific T cell engager (BiTE) antibody against CD19/CD3 for refractory acute lymphoid leukemia. J Hematol Oncol 2015; 8:104. [PMID: 26337639 PMCID: PMC4558758 DOI: 10.1186/s13045-015-0195-4] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Accepted: 08/04/2015] [Indexed: 11/11/2022] Open
Abstract
Targeted therapy has been the forefront of cancer treatment. Cancer immunotherapy is the most recent focus. In addition, novel immunotherapeutics targeting B cell receptor signaling (e.g., ibrutinib), T cell receptor ( e.g., CART19), and NK cells (e.g., AFM13) are being developed. This review summarized the new development in blinatumomab (MT103/MEDI-538), a first-in-class bispecific T engager (BiTE) antibody against CD19/CD3 in patients with relapsed/refractory precursor B cell acute lymphoid leukemia.
Collapse
Affiliation(s)
- Jingjing Wu
- Department of Oncology, The first Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jiaping Fu
- Department of Hematology, Shaoxing People's Hospital, Shaoxing, Zhejiang Province, China
| | - Mingzhi Zhang
- Department of Oncology, The first Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Delong Liu
- Department of Oncology, The first Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China. .,Division of Hematology & Oncology, New York Medical College, Valhalla, NY, 10595, USA.
| |
Collapse
|
4162
|
Iribarren K, Bloy N, Buqué A, Cremer I, Eggermont A, Fridman WH, Fucikova J, Galon J, Špíšek R, Zitvogel L, Kroemer G, Galluzzi L. Trial Watch: Immunostimulation with Toll-like receptor agonists in cancer therapy. Oncoimmunology 2015; 5:e1088631. [PMID: 27141345 DOI: 10.1080/2162402x.2015.1088631] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 08/25/2015] [Indexed: 12/19/2022] Open
Abstract
Accumulating preclinical evidence indicates that Toll-like receptor (TLR) agonists efficiently boost tumor-targeting immune responses (re)initiated by most, if not all, paradigms of anticancer immunotherapy. Moreover, TLR agonists have been successfully employed to ameliorate the efficacy of various chemotherapeutics and targeted anticancer agents, at least in rodent tumor models. So far, only three TLR agonists have been approved by regulatory agencies for use in cancer patients. Moreover, over the past decade, the interest of scientists and clinicians in these immunostimulatory agents has been fluctuating. Here, we summarize recent advances in the preclinical and clinical development of TLR agonists for cancer therapy.
Collapse
Affiliation(s)
- Kristina Iribarren
- INSERM, U1138, Paris, France; Equipe 13, Center de Recherche des Cordeliers, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Norma Bloy
- INSERM, U1138, Paris, France; Gustave Roussy Cancer Campus, Villejuif, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France
| | - Aitziber Buqué
- INSERM, U1138, Paris, France; Gustave Roussy Cancer Campus, Villejuif, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France
| | - Isabelle Cremer
- INSERM, U1138, Paris, France; Equipe 13, Center de Recherche des Cordeliers, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France
| | | | - Wolf Hervé Fridman
- INSERM, U1138, Paris, France; Equipe 13, Center de Recherche des Cordeliers, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France
| | - Jitka Fucikova
- Sotio, Prague, Czech Republic; Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Jérôme Galon
- INSERM, U1138, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Laboratory of Integrative Cancer Immunology, Center de Recherche des Cordeliers, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
| | - Radek Špíšek
- Sotio, Prague, Czech Republic; Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Villejuif, France; INSERM, U1015, CICBT507, Villejuif, France
| | - Guido Kroemer
- INSERM, U1138, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France; Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Lorenzo Galluzzi
- INSERM, U1138, Paris, France; Université Pierre et Marie Curie/Paris VI, Paris, France; Gustave Roussy Cancer Campus, Villejuif, France; Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Center de Recherche des Cordeliers, Paris, France; Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
4163
|
Abstract
Major progress has been made toward our understanding of the programmed death-1/programmed death ligand-1 (PD-1/PD-L1) pathway (referred to as the PD pathway). mAbs are already being used to block the PD pathway to treat human cancers (anti-PD therapy), especially advanced solid tumors. This therapy is based on principles that were discovered through basic research more than a decade ago, but the great potential of this pathway to treat a broad spectrum of advanced human cancers is just now becoming apparent. In this Review, we will briefly review the history and development of anti-PD therapy, from the original benchwork to the most up-to-date clinical results. We will then focus the discussion on three basic principles that define this unique therapeutic approach and highlight how anti-PD therapy is distinct from other immunotherapeutic approaches, namely tumor site immune modulation, targeting tumor-induced immune defects, and repairing ongoing (rather than generating de novo) tumor immunity. We believe that these fundamental principles set the standard for future immunotherapies and will guide our efforts to develop more efficacious and less toxic immune therapeutics to treat human cancers.
Collapse
|
4164
|
Loirat D, Le Tourneau C. Immunothérapie anticancer : les molécules immunomodulatrices en développement clinique. ONCOLOGIE 2015. [DOI: 10.1007/s10269-015-2546-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
4165
|
Endocrinopatías autoinmunitarias inducidas por anticuerpos inmunomoduladores en el tratamiento del cáncer. Med Clin (Barc) 2015; 145:264-8. [DOI: 10.1016/j.medcli.2015.02.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 02/03/2015] [Accepted: 02/07/2015] [Indexed: 01/22/2023]
|
4166
|
Dummer R, Hauschild A, Lindenblatt N, Pentheroudakis G, Keilholz U. Cutaneous melanoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2015; 26 Suppl 5:v126-32. [PMID: 26314774 DOI: 10.1093/annonc/mdv297] [Citation(s) in RCA: 276] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- R Dummer
- Department of Dermatology, University Hospital Zürich, Zürich, Switzerland
| | - A Hauschild
- Department of Dermatology, University Hospital Schleswig-Holstein, Kiel, Germany
| | - N Lindenblatt
- Division of Plastic and Reconstructive Surgery, University Hospital Zürich, Zürich, Switzerland
| | | | - U Keilholz
- Charité Comprehensive Cancer Center, Charité-Universitätsmedizin, Berlin, Germany
| |
Collapse
|
4167
|
Zappasodi R, Merghoub T. Alphavirus-based vaccines in melanoma: rationale and potential improvements in immunotherapeutic combinations. Immunotherapy 2015; 7:981-97. [DOI: 10.2217/imt.15.64] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Immune checkpoint blockade has formally demonstrated the clinical benefit of immunotherapy against melanoma. New immunotherapeutic modalities are currently explored to improve the management of relapsing/refractory patients. Potent antitumor vaccines would have the advantage to promote long-lasting tumor control while limiting autoimmunity. Alphavirus vectors and nonreplicating particles offer versatile platforms to deliver antigen expression and immunize against cancer. They have shown promising preclinical results and initial proof of clinical activity in melanoma. The growing number of clinically available immunomodulatory agents provides a tremendous opportunity to exploit and revisit anticancer vaccines in the setting of powerful immunotherapeutic combinations. Accelerating the evaluation of alphavirus-based vaccines in patients with immune sensitive, but still very deadly malignancies, such as melanoma, is thus extremely important.
Collapse
Affiliation(s)
- Roberta Zappasodi
- Ludwig Collaborative & Swim Across America Laboratory, New York, NY, USA
| | - Taha Merghoub
- Ludwig Collaborative & Swim Across America Laboratory, New York, NY, USA
- Melanoma & Immunotherapeutics Service MSKCC, New York, NY, USA
| |
Collapse
|
4168
|
Borch TH, Donia M, Andersen MH, Svane IM. Reorienting the immune system in the treatment of cancer by using anti-PD-1 and anti-PD-L1 antibodies. Drug Discov Today 2015; 20:1127-34. [DOI: 10.1016/j.drudis.2015.07.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 06/17/2015] [Accepted: 07/09/2015] [Indexed: 02/05/2023]
|
4169
|
PD-L1 expression in small cell lung cancer. Eur J Cancer 2015; 51:1853-5. [DOI: 10.1016/j.ejca.2015.06.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 06/15/2015] [Indexed: 11/20/2022]
|
4170
|
Jamil MO, Lohiya V, Mehta A. Current status of checkpoint inhibitors in lymphoma. Int J Hematol Oncol 2015. [DOI: 10.2217/ijh.15.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cancer immunotherapy is evolving very fast. With understanding of tumor and immune system interactions, two pathways have been identified through which tumor cells evade and escape immune system. Blocking these pathways by monoclonal antibodies has shown promising results in wide variety of tumors. The treatment of lymphomas have been evolving with various new molecules ranging from monoclonal antibodies, antibody–drug conjugates to small molecule inhibitors. Here, we review the activity of immune checkpoint inhibitors in various lymphomas. These agents have shown very promising activity in Hodgkin lymphoma and other B-cell lymphomas in early-phase clinical trials.
Collapse
Affiliation(s)
- Muhammad Omer Jamil
- Division of Hematology & Oncology, Department of Medicine, University of Alabama, 1720 2nd Avenue South, NP2540N, Birmingham, AL 35294, USA
| | - Vipin Lohiya
- Division of Hematology & Oncology, Department of Medicine, University of Alabama, 1720 2nd Avenue South, NP2540N, Birmingham, AL 35294, USA
| | - Amitkumar Mehta
- Division of Hematology & Oncology, Department of Medicine, University of Alabama, 1720 2nd Avenue South, NP2540N, Birmingham, AL 35294, USA
| |
Collapse
|
4171
|
Nicodemus CF. Antibody-based immunotherapy of solid cancers: progress and possibilities. Immunotherapy 2015; 7:923-39. [PMID: 26314410 DOI: 10.2217/imt.15.57] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Monoclonal antibodies remain a primary product option for novel cancer treatment. The properties of an antibody are a function of the antigen specificity and constant region incorporated. The rapid advance in molecular understanding of cancer biology and the host-tumor interaction has defined a new range of targets for antibody development. The clinical success of the checkpoint inhibitors has validated immune modulation and mobilization as a therapeutic approach. Solid cancers are distinguished from hematologic malignancies because the solid tumor stroma contains significant tumor promoting and immune dampening elements less prominent in hematologic cancer. This review highlights how engineered monoclonal antibody products are emerging as potential cornerstones of new more personalized cancer treatment paradigms that target both tumor and the stromal environment.
Collapse
|
4172
|
Mesonero F, López-Sanromán A, Madariaga A, Soria A. [Ipilimumab-induced colitis: A new challenge for gastroenterologists]. GASTROENTEROLOGIA Y HEPATOLOGIA 2015; 39:233-8. [PMID: 26298799 DOI: 10.1016/j.gastrohep.2015.06.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2015] [Revised: 06/18/2015] [Accepted: 06/29/2015] [Indexed: 12/19/2022]
Abstract
Many drugs can produce enterocolitis and they should always be included in the differential diagnosis of this clinical picture. Entities such as antibiotic-associated colitis and neutropenic colitis have been known for some time and recently a new type of drug-induced colitis has emerged due to monoclonal antibodies. Ipimumab is a humanized monoclonal antibody against the CTLA4 molecule that is involved in the maturation and regulation of T lymphocyte activation. This drug causes immune activation and has an immune-mediated antitumor effect with excellent results in tumours such as melanoma. However, several immune-related adverse effects may occur in different organs. The most frequently involved site is the gastrointestinal tract, with adverse effects ranging from mild diarrhoea to colitis with systemic involvement, intestinal perforation, and even death. Although no similarities have been found in the pathogenesis with inflammatory bowel disease, treatments have been used in correlation with its autoimmunological profile: anti-TNF alpha corticosteroids have shown clinical efficacy in moderate to severe disease. However the use of anti-TNF treatment has not been defined and the safety profile is unknown. The inclusion of these new therapies in the treatment of several tumours requires familiarity with these entities and their management should be approached as a new challenge for the gastroenterologist. For that reason, we conducted a review of ipilimumab-induced colitis, evaluating essential features of its symptoms, diagnosis and treatment.
Collapse
Affiliation(s)
- Francisco Mesonero
- Servicio de Gastroenterología y Hepatología, Hospital Universitario Ramón y Cajal, Madrid, España.
| | - Antonio López-Sanromán
- Servicio de Gastroenterología y Hepatología, Hospital Universitario Ramón y Cajal, Madrid, España
| | - Ainhoa Madariaga
- Servicio de Oncología Médica, Hospital Universitario Ramón y Cajal, Madrid, España
| | - Ainara Soria
- Servicio de Oncología Médica, Hospital Universitario Ramón y Cajal, Madrid, España
| |
Collapse
|
4173
|
Shibata T. Current and future molecular profiling of cancer by next-generation sequencing. Jpn J Clin Oncol 2015; 45:895-9. [PMID: 26292697 DOI: 10.1093/jjco/hyv122] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 07/11/2015] [Indexed: 01/18/2023] Open
Abstract
Advances in the new sequencing technologies have enabled us to explore global genetic alterations including driver genes in a wide range of cancers. Concordantly, successes of molecular target therapy promoted the validity of tumor classification based on the combination of targetable genetic abnormalities, and next-generation sequencing-based genetic profiling using target gene capturing or multiplex-polymerase chain reaction has already been tested or adapted in many cancer centers. Driver gene-based classification may be applicable beyond organs, and clinical trials incorporating this genomic information, called as a basket trial, have been executed, although it should be considered that similar therapeutic efficacies against driver mutations are not invariably maintained among different cancer types. Research efforts to identify still missing driver genes in rare cancers, to complete functional annotation of infrequent driver genes, and multiple-layered omics approaches are further expected for better classification of tumor. Emerging clinical interests in the development of immunotherapies postulate a new molecular classification of tumors. Recent studies reported that total number of mutations and the frequent appearance of neo-antigens by protein-coding mutations were associated with a better response, and genetic evaluation of both tumor and host immune system by sequencing is expected to contribute to the personalized immunotherapies in the near future. Lastly intratumoral molecular heterogeneity challenges the current 'static' molecular classification of tumor. For example, dynamic change in clonal constitution within tumor plays an important role in acquired drug resistance. It has been extensively explored whether liquid biopsy-based molecular profiling can resolve currently confronting difficulties.
Collapse
Affiliation(s)
- Tatsuhiro Shibata
- Laboratory of Molecular Medicine, Institute of Medical Science, University of Tokyo, Tokyo Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
4174
|
Abdel-Rahman O, ElHalawani H, Fouad M. Risk of cutaneous toxicities in patients with solid tumors treated with immune checkpoint inhibitors: a meta-analysis. Future Oncol 2015; 11:2471-84. [PMID: 26274495 DOI: 10.2217/fon.15.118] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND We performed a meta-analysis of the risk of cutaneous toxicities associated with immune checkpoint inhibitors. METHODS Eligible studies included randomized trials of patients with solid tumors on immune checkpoint inhibitors (ipilimumab, nivolumab, tremelimumab, pidlizumab and pembrolizumab); describing events of rash, vitiligo and pruritus. RESULTS A total of nine clinical trials were considered eligible for the meta-analysis. The relative risk of all-grade rash, vitiligo and pruritus was 4.06 (95% CI: 3.35-4.91; p < 0.0001), 16.3 (95% CI: 3.21-82.8; p = 0.0008) and 3.4 (95% CI: 2.24-5.16; p < 0.00001), respectively. CONCLUSION Our meta-analysis demonstrates that immune checkpoint inhibitors are associated with an increased risk of all grade skin rash, vitiligo and pruritus. Clinicians should perform regular clinical cutaneous monitoring.
Collapse
Affiliation(s)
- Omar Abdel-Rahman
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hesham ElHalawani
- Clinical Oncology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Mona Fouad
- Medical Microbiology & Immunology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
4175
|
Ribas A. Adaptive Immune Resistance: How Cancer Protects from Immune Attack. Cancer Discov 2015; 5:915-9. [PMID: 26272491 DOI: 10.1158/2159-8290.cd-15-0563] [Citation(s) in RCA: 474] [Impact Index Per Article: 47.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 06/30/2015] [Indexed: 12/24/2022]
Abstract
UNLABELLED Adaptive immune resistance is a process in which the cancer changes its phenotype in response to a cytotoxic or proinflammatory immune response, thereby evading it. This adaptive process is triggered by the specific recognition of cancer cells by T cells, which leads to the production of immune-activating cytokines. Cancers then hijack mechanisms developed to limit inflammatory and immune responses and protect themselves from the T-cell attack. Inhibiting adaptive immune resistance is the mechanistic basis of responses to PD-1 or PD-L1-blocking antibodies, and may be of relevance for the development of other cancer immunotherapy strategies. SIGNIFICANCE Several new immunotherapy strategies to treat cancer are based on inhibiting processes through which cancer adapts and evades from an immune response. Recognizing the specific adaptive resistance mechanisms in each case is likely to allow the personalized development of immunotherapies tailored to block how a particular cancer protects itself from the immune system.
Collapse
Affiliation(s)
- Antoni Ribas
- Division of Hematology-Oncology, Department of Medicine, Jonsson Comprehensive Cancer Center at the University of California, Los Angeles, Los Angeles, California.
| |
Collapse
|
4176
|
Silver DJ, Sinyuk M, Vogelbaum MA, Ahluwalia MS, Lathia JD. The intersection of cancer, cancer stem cells, and the immune system: therapeutic opportunities. Neuro Oncol 2015; 18:153-9. [PMID: 26264894 DOI: 10.1093/neuonc/nov157] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Accepted: 06/17/2015] [Indexed: 01/09/2023] Open
Abstract
During brain neoplasia, malignant cells subjugate the immune system to provide an environment that favors tumor growth. These mechanisms capitalize on tumor-promoting functions of various immune cell types and typically result in suppression of tumor immune rejection. Immunotherapy efforts are underway to disrupt these mechanisms and turn the immune system against developing tumors. While many of these therapies are already in early-stage clinical trials, understanding how these therapies impact various tumor cell populations, including self-renewing cancer stem cells, may help to predict their efficacy and clarify their mechanisms of action. Moreover, interrogating the biology of glioma cell, cancer stem cell, and immune cell interactions may provide additional therapeutic targets to leverage against disease progression. In this review, we begin by highlighting a series of investigations into immune cell-mediated tumor promotion that do not parse the tumor into stem and non-stem components. We then take a closer look at the immune-suppressive mechanisms derived specifically from cancer stem cell interactions with the immune system and end with an update on immunotherapy and cancer stem cell-directed clinical trials in glioblastoma.
Collapse
Affiliation(s)
- Daniel J Silver
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (D.J.S., M.S., J.D.L.); Rose Ella Burkhardt Brain Tumor and Neuro Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., M.S.A., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., M.S.A., J.D.L.); Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (J.D.L.)
| | - Maksim Sinyuk
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (D.J.S., M.S., J.D.L.); Rose Ella Burkhardt Brain Tumor and Neuro Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., M.S.A., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., M.S.A., J.D.L.); Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (J.D.L.)
| | - Michael A Vogelbaum
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (D.J.S., M.S., J.D.L.); Rose Ella Burkhardt Brain Tumor and Neuro Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., M.S.A., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., M.S.A., J.D.L.); Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (J.D.L.)
| | - Manmeet S Ahluwalia
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (D.J.S., M.S., J.D.L.); Rose Ella Burkhardt Brain Tumor and Neuro Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., M.S.A., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., M.S.A., J.D.L.); Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (J.D.L.)
| | - Justin D Lathia
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio (D.J.S., M.S., J.D.L.); Rose Ella Burkhardt Brain Tumor and Neuro Oncology Center, Cleveland Clinic, Cleveland, Ohio (M.A.V., M.S.A., J.D.L.); Case Comprehensive Cancer Center, Cleveland, Ohio (M.A.V., M.S.A., J.D.L.); Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, Ohio (J.D.L.)
| |
Collapse
|
4177
|
Mashima E, Inoue A, Sakuragi Y, Yamaguchi T, Sasaki N, Hara Y, Omoto D, Ohmori S, Haruyama S, Sawada Y, Yoshioka M, Nishio D, Nakamura M. Nivolumab in the treatment of malignant melanoma: review of the literature. Onco Targets Ther 2015; 8:2045-51. [PMID: 26273207 PMCID: PMC4532168 DOI: 10.2147/ott.s62102] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Nivolumab was developed as a monoclonal antibody against programmed death receptor-1, an immune checkpoint inhibitor which negatively regulates T-cell proliferation and activation. Intravenous administration of nivolumab was approved for the treatment of unresectable malignant melanoma in 2014 in Japan. When advanced melanoma patients were treated with nivolumab, median overall survival became longer. Overall survival rate was significantly better in nivolumab-treated melanoma patients than dacarbazine-treated melanoma patients. Nivolumab had an acceptable long-term tolerability profile, with 22% of patients experiencing grade 3 or 4 adverse events related to the drug. Therefore, nivolumab can become an alternative therapy for advanced malignant melanoma.
Collapse
Affiliation(s)
- Emi Mashima
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Akiha Inoue
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yumiko Sakuragi
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Takashi Yamaguchi
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Natsuko Sasaki
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yoko Hara
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Daisuke Omoto
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Shun Ohmori
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Sanehito Haruyama
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Yu Sawada
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Manabu Yoshioka
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Daisuke Nishio
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Motonobu Nakamura
- Department of Dermatology, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
4178
|
Krupka C, Kufer P, Kischel R, Zugmaier G, Lichtenegger FS, Köhnke T, Vick B, Jeremias I, Metzeler KH, Altmann T, Schneider S, Fiegl M, Spiekermann K, Bauerle PA, Hiddemann W, Riethmüller G, Subklewe M. Blockade of the PD-1/PD-L1 axis augments lysis of AML cells by the CD33/CD3 BiTE antibody construct AMG 330: reversing a T-cell-induced immune escape mechanism. Leukemia 2015; 30:484-91. [PMID: 26239198 DOI: 10.1038/leu.2015.214] [Citation(s) in RCA: 203] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 07/06/2015] [Accepted: 07/10/2015] [Indexed: 12/13/2022]
Abstract
Bispecific T-cell engagers (BiTEs) are very effective in recruiting and activating T cells. We tested the cytotoxicity of the CD33/CD3 BiTE antibody construct AMG 330 on primary acute myeloid leukemia (AML) cells ex vivo and characterized parameters contributing to antileukemic cytolytic activity. The E:T ratio and the CD33 expression level significantly influenced lysis kinetics in long-term cultures of primary AML cells (n=38). AMG 330 induced T-cell-mediated proinflammatory conditions, favoring the upregulation of immune checkpoints on target and effector cells. Although not constitutively expressed at the time of primary diagnosis (n=123), PD-L1 was strongly upregulated on primary AML cells upon AMG 330 addition to ex vivo cultures (n=27, P<0.0001). This phenomenon was cytokine-driven as the sole addition of interferon (IFN)-γ and tumor necrosis factor-α also induced expression. Through blockade of the PD-1/PD-L1 interaction, AMG 330-mediated lysis (n=9, P=0.03), T-cell proliferation (n=9, P=0.01) and IFN-γ secretion (n=8, P=0.008) were significantly enhanced. The combinatorial approach was most beneficial in settings of protracted AML cell lysis. Taken together, we have characterized a critical resistance mechanism employed by primary AML cells under AMG 330-mediated proinflammatory conditions. Our results support the evaluation of checkpoint molecules in upcoming clinical trials with AMG 330 to enhance BiTE antibody construct-mediated cytotoxicity.
Collapse
Affiliation(s)
- C Krupka
- Department of Internal Medicine III, LMU-Klinikum der Universität München, Munich, Germany.,Clinical Co-operation Group Immunotherapy at the Helmholtz Zentrum München, Munich, Germany
| | - P Kufer
- AMGEN Research (Munich) GmbH, Munich, Germany
| | - R Kischel
- AMGEN Research (Munich) GmbH, Munich, Germany
| | - G Zugmaier
- AMGEN Research (Munich) GmbH, Munich, Germany
| | - F S Lichtenegger
- Department of Internal Medicine III, LMU-Klinikum der Universität München, Munich, Germany.,Clinical Co-operation Group Immunotherapy at the Helmholtz Zentrum München, Munich, Germany
| | - T Köhnke
- Department of Internal Medicine III, LMU-Klinikum der Universität München, Munich, Germany.,Clinical Co-operation Group Immunotherapy at the Helmholtz Zentrum München, Munich, Germany
| | - B Vick
- Department of Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - I Jeremias
- Department of Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Oncology, Dr von Haunersches Kinderspital, Ludwig Maximilians-Universität (LMU), Munich, Germany
| | - K H Metzeler
- Department of Internal Medicine III, LMU-Klinikum der Universität München, Munich, Germany
| | - T Altmann
- Department of Internal Medicine III, LMU-Klinikum der Universität München, Munich, Germany.,Clinical Co-operation Group Immunotherapy at the Helmholtz Zentrum München, Munich, Germany
| | - S Schneider
- Department of Internal Medicine III, LMU-Klinikum der Universität München, Munich, Germany
| | - M Fiegl
- Department of Internal Medicine III, LMU-Klinikum der Universität München, Munich, Germany
| | - K Spiekermann
- Department of Internal Medicine III, LMU-Klinikum der Universität München, Munich, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - P A Bauerle
- AMGEN Research (Munich) GmbH, Munich, Germany
| | - W Hiddemann
- Department of Internal Medicine III, LMU-Klinikum der Universität München, Munich, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - G Riethmüller
- Institute for Immunology, Ludwig-Maximilians-Universität, Munich, Germany
| | - M Subklewe
- Department of Internal Medicine III, LMU-Klinikum der Universität München, Munich, Germany.,Clinical Co-operation Group Immunotherapy at the Helmholtz Zentrum München, Munich, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
4179
|
Kim KB. Nivolumab in the treatment of advanced melanoma. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1063416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
4180
|
Bowyer S, Lorigan P. The place of PD-1 inhibitors in melanoma management. Lancet Oncol 2015; 16:873-4. [DOI: 10.1016/s1470-2045(15)00094-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 06/15/2015] [Indexed: 10/23/2022]
|
4181
|
Berghoff AS, Preusser M. The future of targeted therapies for brain metastases. Future Oncol 2015; 11:2315-27. [DOI: 10.2217/fon.15.127] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Brain metastases (BM) are an increasing challenge in the management of patients with advanced cancer. Treatment options for BM are limited and mainly focus on the application of local therapies. Systemic therapies including targeted therapies are only poorly investigated, as patients with BM were frequently excluded from clinical trials. Several targeted therapies have shown promising activity in patients with BM. In the present review we discuss existing and emerging targeted therapies for the most frequent BM primary tumor types. We focus on challenges in the conduction of clinical trials on targeted therapies in BM patients such as patient selection, combination with radiotherapy, the obstacles of the blood–brain barrier and the definition of study end points.
Collapse
Affiliation(s)
- Anna S Berghoff
- Department for Medicine I, Comprehensive Cancer Center Central Nervous System Unit (CCC-CNS), Clinical Division of Oncology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center – CNS Tumors Unit, Medical University of Vienna, Vienna, Austria
| | - Matthias Preusser
- Department for Medicine I, Comprehensive Cancer Center Central Nervous System Unit (CCC-CNS), Clinical Division of Oncology, Medical University of Vienna, Vienna, Austria
- Comprehensive Cancer Center – CNS Tumors Unit, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
4182
|
Long GV, Stroyakovskiy D, Gogas H, Levchenko E, de Braud F, Larkin J, Garbe C, Jouary T, Hauschild A, Grob JJ, Chiarion-Sileni V, Lebbe C, Mandalà M, Millward M, Arance A, Bondarenko I, Haanen JBAG, Hansson J, Utikal J, Ferraresi V, Kovalenko N, Mohr P, Probachai V, Schadendorf D, Nathan P, Robert C, Ribas A, DeMarini DJ, Irani JG, Swann S, Legos JJ, Jin F, Mookerjee B, Flaherty K. Dabrafenib and trametinib versus dabrafenib and placebo for Val600 BRAF-mutant melanoma: a multicentre, double-blind, phase 3 randomised controlled trial. Lancet 2015; 386:444-51. [PMID: 26037941 DOI: 10.1016/s0140-6736(15)60898-4] [Citation(s) in RCA: 1025] [Impact Index Per Article: 102.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Previously, a study of ours showed that the combination of dabrafenib and trametinib improves progression-free survival compared with dabrafenib and placebo in patients with BRAF Val600Lys/Glu mutation-positive metastatic melanoma. The study was continued to assess the secondary endpoint of overall survival, which we report in this Article. METHODS We did this double-blind phase 3 study at 113 sites in 14 countries. We enrolled previously untreated patients with BRAF Val600Glu or Val600Lys mutation-positive unresectable stage IIIC or stage IV melanoma. Participants were computer-randomised (1:1) to receive a combination of dabrafenib (150 mg orally twice daily) and trametinib (2 mg orally once daily), or dabrafenib and placebo. The primary endpoint was progression-free survival and overall survival was a secondary endpoint. This study is registered with ClinicalTrials.gov, number NCT01584648. FINDINGS Between May 4, 2012, and Nov 30, 2012, we screened 947 patients for eligibility, of whom 423 were randomly assigned to receive dabrafenib and trametinib (n=211) or dabrafenib only (n=212). The final data cutoff was Jan 12, 2015, at which time 222 patients had died. Median overall survival was 25·1 months (95% CI 19·2-not reached) in the dabrafenib and trametinib group versus 18·7 months (15·2-23·7) in the dabrafenib only group (hazard ratio [HR] 0·71, 95% CI 0·55-0·92; p=0·0107). Overall survival was 74% at 1 year and 51% at 2 years in the dabrafenib and trametinib group versus 68% and 42%, respectively, in the dabrafenib only group. Based on 301 events, median progression-free survival was 11·0 months (95% CI 8·0-13·9) in the dabrafenib and trametinib group and 8·8 months (5·9-9·3) in the dabrafenib only group (HR 0·67, 95% CI 0·53-0·84; p=0·0004; unadjusted for multiple testing). Treatment-related adverse events occurred in 181 (87%) of 209 patients in the dabrafenib and trametinib group and 189 (90%) of 211 patients in the dabrafenib only group; the most common was pyrexia (108 patients, 52%) in the dabrafenib and trametinib group, and hyperkeratosis (70 patients, 33%) in the dabrafenib only group. Grade 3 or 4 adverse events occurred in 67 (32%) patients in the dabrafenib and trametinib group and 66 (31%) patients in the dabrafenib only group. INTERPRETATION The improvement in overall survival establishes the combination of dabrafenib and trametinib as the standard targeted treatment for BRAF Val600 mutation-positive melanoma. Studies assessing dabrafenib and trametinib in combination with immunotherapies are ongoing. FUNDING GlaxoSmithKline.
Collapse
Affiliation(s)
- Georgina V Long
- Melanoma Institute Australia, University of Sydney, Sydney, NSW, Australia; Mater Hospital, Sydney, NSW, Australia.
| | | | - Helen Gogas
- Department of Medicine, University of Athens, Medical School, Athens, Greece
| | | | | | | | - Claus Garbe
- Department of Dermatology, University Hospital Tuebingen, Tuebingen, Germany
| | - Thomas Jouary
- Department of Dermatology, Hôpital Saint André, CHU Bordeaux, Bordeaux, France
| | | | - Jean-Jacques Grob
- Aix-Marseille University and APHM Hospital CHU Timone, Marseille, France
| | - Vanna Chiarion-Sileni
- Melanoma and Oesophageal Oncology Unit, Veneto Oncology Institute-IRCCS, Padua, Italy
| | - Celeste Lebbe
- APHP Dermatology CIC Hôpital Saint Louis, University Paris Diderot, INSERM U976, Paris, France
| | - Mario Mandalà
- Papa Giovanni XXIII Cancer Center Hospital, Bergamo, Italy
| | - Michael Millward
- Sir Charles Gairdner Hospital, Hospital Avenue, Perth, WA, Australia
| | - Ana Arance
- Department of Medical Oncology, Hospital Clinic and Translational Genomics and Targeted Therapeutics in Solid Tumors, Barcelona, Spain
| | - Igor Bondarenko
- Dnepropetrovsk State Medical Academy, Dnepropetrovsk, Ukraine
| | | | - Johan Hansson
- Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Jochen Utikal
- University Medical Center Mannheim, Heidelberg University, Mannheim, Germany; German Cancer Research Center, Heidelberg, Germany
| | | | | | | | - Volodymr Probachai
- Dnipropetrovsk Clinical Oncology Center of Dnipropetrovsk State Council, Dnipropetrovsk, Ukraine
| | | | | | - Caroline Robert
- Gustave Roussy, Villejuif-Paris-Sud, France; Paris Sud University, Le Kremlin Bicêtre, France
| | - Antoni Ribas
- David Geffen School of Medicine, UCLA, Los Angeles, CA, USA
| | | | | | - Suzanne Swann
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | | - Fan Jin
- Merck & Co, Kenilworth, NJ, USA
| | | | - Keith Flaherty
- Massachusetts General Hospital Cancer Center, Boston MA, USA
| |
Collapse
|
4183
|
Yélamos O, Gerami P. Predicting the outcome of melanoma: can we tell the future of a patient's melanoma? Melanoma Manag 2015; 2:217-224. [PMID: 30190851 PMCID: PMC6094684 DOI: 10.2217/mmt.15.15] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cutaneous melanoma is responsible for the greatest number of skin cancer related deaths. For many years there were few therapeutic options. However, in the last years a number of new therapeutic options have emerged showing improved survival rates for advanced melanoma patients. A significant question based on these findings is whether identification and treatment of patients with biologically aggressive melanomas at an earlier clinical stage offer an opportunity for even greater improvement in overall survival. In this review, we will discuss the recent advancements in molecular strategies beyond traditional staging to identify biologically aggressive melanomas, and which are their implications in terms of predicting the prognosis of patients with melanoma.
Collapse
Affiliation(s)
- Oriol Yélamos
- Department of Dermatology, Feinberg School of Medicine, The Robert H Lurie Cancer Center, Northwestern University, 676 N. St Clair Street, Suite 1765, Chicago, IL 60611, USA
| | - Pedram Gerami
- Department of Dermatology, Feinberg School of Medicine, The Robert H Lurie Cancer Center, Northwestern University, 676 N. St Clair Street, Suite 1765, Chicago, IL 60611, USA
| |
Collapse
|
4184
|
Abstract
The potential to harness the power of the immune system and effectively treat patients with metastatic melanoma is finally being realized with the advent of immune checkpoint inhibitors. These new therapies herald a new era in the treatment of melanoma with the potential to produce very durable responses and possible cure for a subset of patients, though bring with them challenges including novel toxicities and nonconventional response patterns. This article reviews the currently available immune checkpoint inhibitors, potential biomarkers to predict response and promising investigational approaches including combination therapies.
Collapse
Affiliation(s)
- Adam J Cooper
- Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, NSW, Australia
- School of Medicine, The University of Sydney, Sydney, NSW, Australia
- Melanoma Institute Australia, Sydney, NSW, Australia
| | - Matteo S Carlino
- Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, NSW, Australia
- School of Medicine, The University of Sydney, Sydney, NSW, Australia
- Melanoma Institute Australia, Sydney, NSW, Australia
- Westmead Institute for Cancer Research, University of Sydney at the Westmead Millennium Institute, Westmead, NSW, Australia
| | - Richard F Kefford
- Crown Princess Mary Cancer Centre, Westmead Hospital, Sydney, NSW, Australia
- School of Medicine, The University of Sydney, Sydney, NSW, Australia
- Melanoma Institute Australia, Sydney, NSW, Australia
- Westmead Institute for Cancer Research, University of Sydney at the Westmead Millennium Institute, Westmead, NSW, Australia
- Department of Clinical Medicine, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
4185
|
Affiliation(s)
- Keiran S M Smalley
- Department of Tumor Biology, The Moffitt Cancer Center, Tampa, FL 33612, USA; Department of Cutaneous Oncology, The Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Vernon K Sondak
- Department of Cutaneous Oncology, The Moffitt Cancer Center, Tampa, FL 33612, USA.
| |
Collapse
|
4186
|
Karsy M, Guan J, Sivakumar W, Neil JA, Schmidt MH, Mahan MA. The genetic basis of intradural spinal tumors and its impact on clinical treatment. Neurosurg Focus 2015; 39:E3. [DOI: 10.3171/2015.5.focus15143] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Genetic alterations in the cells of intradural spinal tumors can have a significant impact on the treatment options, counseling, and prognosis for patients. Although surgery is the primary therapy for most intradural tumors, radiochemothera-peutic modalities and targeted interventions play an ever-evolving role in treating aggressive cancers and in addressing cancer recurrence in long-term survivors. Recent studies have helped delineate specific genetic and molecular differences between intradural spinal tumors and their intracranial counterparts and have also identified significant variation in therapeutic effects on these tumors. This review discusses the genetic and molecular alterations in the most common intradural spinal tumors in both adult and pediatrie patients, including nerve sheath tumors (that is, neurofibroma and schwannoma), meningioma, ependymoma, astrocytoma (that is, low-grade glioma, anaplastic astrocytoma, and glioblastoma), hemangioblastoma, and medulloblastoma. It also examines the genetics of metastatic tumors to the spinal cord, arising either from the CNS or from systemic sources. Importantly, the impact of this knowledge on therapeutic options and its application to clinical practice are discussed.
Collapse
|
4187
|
González-Cao M, Karachaliou N, Viteri S, Morales-Espinosa D, Teixidó C, Sánchez Ruiz J, Molina-Vila MÁ, Santarpia M, Rosell R. Targeting PD-1/PD-L1 in lung cancer: current perspectives. LUNG CANCER (AUCKLAND, N.Z.) 2015; 6:55-70. [PMID: 28210151 PMCID: PMC5217517 DOI: 10.2147/lctt.s55176] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Increased understanding of tumor immunology has led to the development of effective immunotherapy treatments. One of the most important advances in this field has been due to pharmacological design of antibodies against immune checkpoint inhibitors. Anti-PD-1/PD-L1 antibodies are currently in advanced phases of clinical development for several tumors, including lung cancer. Results from Phase I-III trials with anti-PD-1/PD-L1 antibodies in non-small-cell lung cancer have demonstrated response rates of around 20% (range, 16%-50%). More importantly, responses are long-lasting (median duration of response, 18 months) and fast (50% of responses are detected at time of first tumor evaluation) with very low grade 3-4 toxicity (less than 5%). Recently, the anti-PD-1 antibody pembrolizumab received US Food and Drug Administration (FDA) breakthrough therapy designation for treatment of non-small-cell lung cancer, supported by data from a Phase Ib trial. Another anti-PD-1 antibody, nivolumab, has also been approved for lung cancer based on survival advantage demonstrated in recently released data from a Phase III trial in squamous cell lung cancer.
Collapse
Affiliation(s)
- María González-Cao
- Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Niki Karachaliou
- Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Santiago Viteri
- Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Daniela Morales-Espinosa
- Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain
| | | | | | | | - Mariacarmela Santarpia
- Medical Oncology Unit, Human Pathology Department, University of Messina, Messina, Italy
| | - Rafael Rosell
- Translational Cancer Research Unit, Instituto Oncológico Dr Rosell, Quirón Dexeus University Hospital, Barcelona, Spain
- Pangaea Biotech SL, Barcelona, Spain
- Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Germans Trias i Pujol Health Sciences Institute and Hospital, Campus Can Ruti, Badalona, Barcelona, Spain
- Fundación Molecular Oncology Research, Barcelona, Spain
| |
Collapse
|
4188
|
Abstract
PURPOSE AND DESIGN In recent years, there have been dramatic improvements in the diagnosis and treatment of patients with melanoma. The development of molecular markers and associated targeted therapies have given new hope to subsets of patients with advanced disease. Here we discuss the most important advances in molecular targeted therapy and how these developments are likely to affect the practice of the clinical surgeon. RESULTS AND CONCLUSIONS Germ-line and somatic mutations are common in melanoma and provide prognostic information that can now be harnessed to provide a more personalized approach to cancer treatment. BRAF mutation at the V600 position is the most commonly identified mutation in patients with melanoma. Treatment with targeted inhibitors in patients with BRAF-mutant melanoma has afforded dramatic responses in about half of selected patients. Unfortunately, disease control is not durable and recurrences are common. We predict an increasing role for the surgeon in the multidisciplinary treatment of patients with metastatic disease, as well as a role for molecular profiling in patients with high-risk early stage disease. Further, we are only beginning to understand the prognostic significance of various gene mutations in patients with melanoma.
Collapse
Affiliation(s)
- Danielle K DePeralta
- Division of Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Genevieve M Boland
- Division of Surgical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4189
|
Faghfuri E, Faramarzi MA, Nikfar S, Abdollahi M. Nivolumab and pembrolizumab as immune-modulating monoclonal antibodies targeting the PD-1 receptor to treat melanoma. Expert Rev Anticancer Ther 2015; 15:981-993. [PMID: 26313415 DOI: 10.1586/14737140.2015.1074862] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Malignant melanoma is an important issue in oncology due to its high incidence, high mortality, and resistance to systemic therapy; however, targeted immunotherapy has noticeably improved the survival rates of melanoma patients. Promising targeted immunotherapies for malignant melanoma include the blockade of immune checkpoints with antibodies targeting cytotoxic T lymphocyte-associated antigen 4 and the programmed cell death protein 1 pathway. The US FDA-approved antibody ipilimumab targets cytotoxic T lymphocyte-associated antigen 4; however, it was limited by toxicity and a low response. Nivolumab and pembrolizumab (formerly lambrolizumab), the two FDA-approved anti-programmed death-1 monoclonal antibodies, show highly durable response rates and long-term safety, validating the importance of the programmed cell death protein 1 pathway blockade for treatment of malignant melanoma.
Collapse
Affiliation(s)
- Elnaz Faghfuri
- a 1 Department of Pharmaceutical Biotechnology and Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | |
Collapse
|
4190
|
Guo Z, Wang H, Meng F, Li J, Zhang S. Combined Trabectedin and anti-PD1 antibody produces a synergistic antitumor effect in a murine model of ovarian cancer. J Transl Med 2015. [PMID: 26219551 PMCID: PMC4517526 DOI: 10.1186/s12967-015-0613-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Monoclonal antibodies (mAb) that block programmed death (PD)-1 signaling pathway hold great potential as a novel cancer immunotherapy. Recent evidence suggests that combining with conventional, targeted or other immunotherapies, these mAb can induce synergistic antitumor responses. In this study, we investigated whether Trabectedin (ET-743), a novel anticancer agent currently used for treating relapsed ovarian cancer, can synergize with anti (α)-PD-1 mAb to increase antitumor activity in the murine ID8 ovarian cancer model. METHODS Mice with established peritoneal ID8 tumor were treated with either single or combined Trabectedin and α-PD-1 mAb, their overall survival was recorded; tumor-associated immune cells and immune gene expression in tumors from treated mice were analyzed by flow cytometry and quantitative RT-PCR, respectively, and antigen-specific immunity of effector CD8(+) T cells was evaluated by ELISA and cytotoxicity assay. In addition, the effect of Trabectedin on tumoral PD-L1 expression was analyzed by both flow cytometry and immunofluorescence staining. RESULTS Though single treatment showed a modest antitumor effect in mice bearing 10-day-established ID8 tumor, combined Trabectedin and α-PD-1 mAb treatment induced a strong antitumor immune response, leading to a significant tumor regression with half of mice tumor-free 90 days after tumor inoculation. Mechanistic investigation revealed that combination treatment induces a systemic tumor-specific immunity with an indispensable role of both CD4(+) and CD8(+) T cells, and effector CD8(+) T cells exhibited the antigen-specific cytokine secretion and cytotoxicity upon tumor antigen stimulation; additionally, combination treatment increased the IFN-γ-producing effector T cells and decreased the immunosuppressive cells in peritoneal cavity; accordingly, it enhanced the expression of Th1-associated immune-stimulating genes while reducing the transcription of regulatory/suppressive immune genes, reshaping tumor microenvironment from a immunosuppressive to a stimulatory state. Finally, in vivo Trabectedin treatment has been shown to induce IFN-γ-dependent PD-L1 expression within tumor, possibly constituting a mechanistic basis for its synergistic antitumor effect with α-PD-1 mAb therapy. CONCLUSION This study provides the evidence that α-PD-1 mAb can produce a synergistic antitumor efficacy when combined with Trabectedin, a clinically available anticancer agent, supporting a direct translation of this combination strategy in clinic for the treatment of ovarian cancer.
Collapse
Affiliation(s)
- Zhiqiang Guo
- Department of Gynecology and Obstetrics, Shengjing Hospital, China Medical University, Shenyang, 110004, China.
| | - Haolin Wang
- Department of Acute Abdominal Surgery, The First Hospital of Dalian Medical University, Dalian, 116044, China.
| | - Fandong Meng
- Molecular Oncology Department of Cancer Research Institution, The First Hospital of China Medical University, Shenyang, 110004, China.
| | - Jie Li
- Department of Gynecology and Obstetrics, Shengjing Hospital, China Medical University, Shenyang, 110004, China.
| | - Shulan Zhang
- Department of Gynecology and Obstetrics, Shengjing Hospital, China Medical University, Shenyang, 110004, China.
| |
Collapse
|
4191
|
Marzuka A, Huang L, Theodosakis N, Bosenberg M. Melanoma Treatments: Advances and Mechanisms. J Cell Physiol 2015; 230:2626-33. [DOI: 10.1002/jcp.25019] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Accepted: 04/14/2015] [Indexed: 02/03/2023]
Affiliation(s)
- Alexander Marzuka
- Department of Dermatology; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
- Department of Internal Medicine; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
| | - Laura Huang
- Department of Dermatology; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
- Department of Internal Medicine; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
| | - Nicholas Theodosakis
- Department of Pathology; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
| | - Marcus Bosenberg
- Department of Dermatology; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
- Department of Pathology; Yale-New Haven Hospital; Yale School of Medicine; New Haven Connecticut
| |
Collapse
|
4192
|
Ngiow SF, Young A, Jacquelot N, Yamazaki T, Enot D, Zitvogel L, Smyth MJ. A Threshold Level of Intratumor CD8+ T-cell PD1 Expression Dictates Therapeutic Response to Anti-PD1. Cancer Res 2015. [PMID: 26208901 DOI: 10.1158/0008-5472.can-15-1082] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Despite successes, thus far, a significant proportion of the patients treated with anti-PD1 antibodies have failed to respond. We use mouse tumor models of anti-PD1 sensitivity and resistance and flow cytometry to assess tumor-infiltrating immune cells immediately after therapy. We demonstrate that the expression levels of T-cell PD1 (PD1(lo)), myeloid, and T-cell PDL1 (PDL1(hi)) in the tumor microenvironment inversely correlate and dictate the efficacy of anti-PD1 mAb and function of intratumor CD8(+) T cells. In sensitive tumors, we reveal a threshold for PD1 downregulation on tumor-infiltrating CD8(+) T cells below which the release of adaptive immune resistance is achieved. In contrast, PD1(hi) T cells in resistant tumors fail to be rescued by anti-PD1 therapy and remain dysfunctional unless intratumor PDL1(lo) immune cells are targeted. Intratumor Tregs are partly responsible for the development of anti-PD1-resistant tumors and PD1(hi) CD8(+) T cells. Our analyses provide a framework to interrogate intratumor CD8(+) T-cell PD1 and immune PDL1 levels and response in human cancer.
Collapse
MESH Headings
- Adaptive Immunity
- Adenocarcinoma/immunology
- Adenocarcinoma/therapy
- Animals
- Antibodies, Monoclonal/therapeutic use
- B7-H1 Antigen/physiology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Colonic Neoplasms/immunology
- Colonic Neoplasms/therapy
- Down-Regulation
- Female
- Gene Expression Regulation, Neoplastic/immunology
- Immunization, Passive
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Mammary Neoplasms, Experimental/immunology
- Mammary Neoplasms, Experimental/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- Myeloid Cells/immunology
- Myeloid Cells/metabolism
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neoplasm Proteins/physiology
- Programmed Cell Death 1 Receptor/biosynthesis
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/physiology
- T-Lymphocytes, Regulatory/immunology
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Shin Foong Ngiow
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia. School of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Arabella Young
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia. School of Medicine, University of Queensland, Herston, Queensland, Australia
| | - Nicolas Jacquelot
- INSERM U1015, Villejuif, France. Institut de Cancérologie Gustave Roussy, Villejuif, France. Equipe labellisée Ligue contre le Cancer, University of Paris Sud XI, Villejuif, France. Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France
| | - Takahiro Yamazaki
- INSERM U1015, Villejuif, France. Institut de Cancérologie Gustave Roussy, Villejuif, France. Equipe labellisée Ligue contre le Cancer, University of Paris Sud XI, Villejuif, France. Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France
| | - David Enot
- INSERM U1015, Villejuif, France. Institut de Cancérologie Gustave Roussy, Villejuif, France. Equipe labellisée Ligue contre le Cancer, University of Paris Sud XI, Villejuif, France. Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France. Metabolomics and Cell Biology Platforms, Institut de Cancerologie Gustave Roussy, Villejuif, France
| | - Laurence Zitvogel
- INSERM U1015, Villejuif, France. Institut de Cancérologie Gustave Roussy, Villejuif, France. Equipe labellisée Ligue contre le Cancer, University of Paris Sud XI, Villejuif, France. Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France
| | - Mark J Smyth
- Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia. School of Medicine, University of Queensland, Herston, Queensland, Australia.
| |
Collapse
|
4193
|
Kranz LM, Birtel M, Krienke C, Grunwitz C, Petschenka J, Reuter KC, van de Roemer N, Vascotto F, Vormehr M, Kreiter S, Diken M. CIMT 2015: The right patient for the right therapy - Report on the 13th annual meeting of the Association for Cancer Immunotherapy. Hum Vaccin Immunother 2015; 12:213-21. [PMID: 26186022 PMCID: PMC4962731 DOI: 10.1080/21645515.2015.1068485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 06/29/2015] [Indexed: 12/22/2022] Open
Affiliation(s)
- Lena M Kranz
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
- Research Center for Immunotherapy (FZI); University Medical Center; Johannes Gutenberg University; Mainz, Germany
| | - Matthias Birtel
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
| | - Christina Krienke
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
- Research Center for Immunotherapy (FZI); University Medical Center; Johannes Gutenberg University; Mainz, Germany
| | - Christian Grunwitz
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
- BioNTech RNA Pharmaceuticals GmbH; Mainz, Germany
| | - Jutta Petschenka
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
| | | | - Niels van de Roemer
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
- Research Center for Immunotherapy (FZI); University Medical Center; Johannes Gutenberg University; Mainz, Germany
| | - Fulvia Vascotto
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
| | - Mathias Vormehr
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
- BioNTech RNA Pharmaceuticals GmbH; Mainz, Germany
| | - Sebastian Kreiter
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
| | - Mustafa Diken
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg University Mainz gGmbH; Mainz, Germany
| |
Collapse
|
4194
|
Santarpia M, Karachaliou N. Tumor immune microenvironment characterization and response to anti-PD-1 therapy. Cancer Biol Med 2015; 12:74-8. [PMID: 26175922 PMCID: PMC4493379 DOI: 10.7497/j.issn.2095-3941.2015.0022] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 05/08/2015] [Indexed: 12/16/2022] Open
Affiliation(s)
- Mariacarmela Santarpia
- 1 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 2 Translational Research Unit, Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, Barcelona 08028, Spain
| | - Niki Karachaliou
- 1 Medical Oncology Unit, Human Pathology Department, University of Messina, Messina 98122, Italy ; 2 Translational Research Unit, Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, Barcelona 08028, Spain
| |
Collapse
|
4195
|
Ascierto ML, Melero I, Ascierto PA. Melanoma: From Incurable Beast to a Curable Bet. The Success of Immunotherapy. Front Oncol 2015; 5:152. [PMID: 26217587 PMCID: PMC4500097 DOI: 10.3389/fonc.2015.00152] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 06/22/2015] [Indexed: 01/22/2023] Open
Abstract
After Coley's observation in 1891 of tumor regression in a patient who developed a postoperative infection, the field of immunotherapy is finally reborn. Avoiding immune destruction is now considered a hallmark of cancer, and the immunotherapy arena has exploded with the recent advances demonstrating an improvement in survival and a durability of response in patients with different cancer types, which translates into improved overall survival benefit. Here, we provide an overview of the main immune-oncology treatment strategies that, either alone or in combination, are undergoing clinical development. Namely, we will refer to those immunotherapeutic strategies that include adoptive transfer of ex vivo activated T cells, immunomodulatory monoclonal antibodies, and cancer vaccines. Our major focus will be to describe these approaches in melanoma, a cancer type transformed by immunotherapy into a potentially curable disease.
Collapse
Affiliation(s)
| | - Ignacio Melero
- Department of Oncology, Centro de Investigación Médica Aplicada (CIMA), Clinica Universidad de Navarra, Pamplona, Spain
| | - Paolo Antonio Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale Tumori Fondazione “G. Pascale”, Napoli, Italy
| |
Collapse
|
4196
|
Abstract
The immune system plays a vital role in regulating tumor growth, and the oncology community has witnessed an exciting resurgence in clinical research to develop effective immunotherapeutic strategies. The utility of these strategies in advanced melanoma has been at the forefront of these developments. In particular, blockade of programmed cell death protein 1 (PD-1) in advanced melanoma has proven to be a most promising new anticancer strategy. Pembrolizumab is a humanized IgG4 anti-PD-1 antibody that exerts its anti-tumor effect through blocking the interaction of the immune inhibitory molecule PD-1 with its ligands. Its effect has been most convincingly demonstrated in the setting of advanced melanoma, with growing evidence of clinical responses across a broad spectrum of other solid and hematological malignancies.
Collapse
Affiliation(s)
- Sanjeev Srinivas Kumar
- a Department of Medical Oncology, Chris O'Brien Lifehouse, 119-143 Missenden Road, Camperdown NSW 2050, PO BOX M33 Missenden Road NSW 2050, Australia
| | | |
Collapse
|
4197
|
Chinai JM, Janakiram M, Chen F, Chen W, Kaplan M, Zang X. New immunotherapies targeting the PD-1 pathway. Trends Pharmacol Sci 2015; 36:587-95. [PMID: 26162965 DOI: 10.1016/j.tips.2015.06.005] [Citation(s) in RCA: 141] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/15/2015] [Accepted: 06/17/2015] [Indexed: 12/22/2022]
Abstract
Ligands from the B7 family bind to receptors of the CD28 family, which regulate early T cell activation in lymphoid organs and control inflammation and autoimmunity in peripheral tissues. Programmed death-1 (PD-1), a member of the CD28 family, is an inhibitory receptor on T cells and is responsible for their dysfunction in infectious diseases and cancers. The complex mechanisms controlling the expression and signaling of PD-1 and programmed death ligand 1 (PD-L1) are emerging. Recently completed and ongoing clinical trials that target these molecules have shown remarkable success by generating durable clinical responses in some cancer patients. In chronic viral infections, preclinical data reveal that targeting PD-1 and its ligands can improve T cell responses and virus clearance. There is also promise in stimulating this pathway for the treatment of autoimmune and inflammatory disorders.
Collapse
Affiliation(s)
- Jordan M Chinai
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Murali Janakiram
- Department of Oncology, Montefiore Medical Center, New York, NY 10467, USA
| | - Fuxiang Chen
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Wantao Chen
- Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Mark Kaplan
- Centers for Therapeutic Innovation, Pfizer Inc., New York, NY 10016, USA
| | - Xingxing Zang
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, New York, NY 10461, USA; Department of Oncology, Montefiore Medical Center, New York, NY 10467, USA.
| |
Collapse
|
4198
|
Ascierto PA, Marincola FM, Atkins MB. What's new in melanoma? Combination! J Transl Med 2015; 13:213. [PMID: 26141621 PMCID: PMC4491255 DOI: 10.1186/s12967-015-0582-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 06/25/2015] [Indexed: 01/22/2023] Open
Abstract
Melanoma was again a focus of attention at the 2015 American Society of Clinical Oncology (ASCO) Annual Meeting, in particular the use of combination treatment strategies involving immunotherapies and/or targeted agents. New data on targeted therapies confirmed previous findings, with combined BRAF inhibitor (vemurafenib) plus MEK inhibitor (cobimetinib) improving progression-free survival (PFS) compared to vemurafenib monotherapy in patients with BRAFV600 mutation-positive tumors (CoBRIM trial). Positive results were also seen with combined dabrafenib and trametinib in patients with BRAF V600E/K metastatic melanoma and encorafenib plus binimetinib in BRAFV600-mutant cutaneous melanoma. Even more interesting news centered on the use of combination immunotherapy, in particular the randomized, double-blind CheckMate 067 study in which median PFS with nivolumab plus ipilimumab was 11.5 months, compared to 2.9 months with ipilimumab alone (HR 0.42) and 6.9 months with nivolumab alone (HR 0.57). Of interest, in patients with ≥5% PD-L1 expression, median PFS was 14 months with the combination or with nivolumab alone compared with 3.9 months in the ipilimumab group, while in the PD-L1 negative cohort, the combination remained superior to both monotherapies. Given that combination therapy was accompanied by a high occurrence of side-effects, this raises the suggestion that combination therapy might be reserved for PD-L1 negative patients only, with PD-L1 positive patients achieving the same benefit from nivolumab monotherapy. However, overall survival data are awaited and the equivalence of single agent to the combination remains unconvincing. Interesting data were also reported on the combination of T-VEC (talimogene laherparepvec) with ipilimumab, and the anti-PD-1 agent MEDI4736 (durvolumab) combined with dabrafenib plus trametinib. Emerging data also suggested that predictive markers based on immunoprofiling and mismatch repair deficiency may be of clinical use. In conclusion, the use of combination approaches to treat patients with melanoma, as well as other cancers, is no longer a just a wish for the future but is today a clinical reality with a rapidly growing evidence-base. Moreover, the most exciting consideration is that this is far from the end of the story, but rather a fantastic introduction.
Collapse
Affiliation(s)
- Paolo A Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Innovative Therapy, Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione G. Pascale", Via Mariano Semmola, 80131, Naples, Italy.
| | | | - Michael B Atkins
- Oncology and Medicine, Georgetown-Lombardi Comprehensive Cancer Center, Georgetown University School of Medicine, 3970 Reservoir Rd NW, Washington, DC, 20057, USA.
| |
Collapse
|
4199
|
Fang B, Mehran RJ, Heymach JV, Swisher SG. Predictive biomarkers in precision medicine and drug development against lung cancer. CHINESE JOURNAL OF CANCER 2015; 34:295-309. [PMID: 26134262 PMCID: PMC4593363 DOI: 10.1186/s40880-015-0028-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 06/04/2015] [Indexed: 02/06/2023]
Abstract
The molecular characterization of various cancers has shown that cancers with the same origins, histopathologic diagnoses, and clinical stages can be highly heterogeneous in their genetic and epigenetic alterations that cause tumorigenesis. A number of cancer driver genes with functional abnormalities that trigger malignant transformation and that are required for the survival of cancer cells have been identified. Therapeutic agents targeting some of these cancer drivers have been successfully developed, resulting in substantial improvements in clinical symptom amelioration and outcomes in a subset of cancer patients. However, because such therapeutic drugs often benefit only a limited number of patients, the successes of clinical development and applications rely on the ability to identify those patients who are sensitive to the targeted therapies. Thus, biomarkers that can predict treatment responses are critical for the success of precision therapy for cancer patients and of anticancer drug development. This review discusses the molecular heterogeneity of lung cancer pathogenesis; predictive biomarkers for precision medicine in lung cancer therapy with drugs targeting epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), c-ros oncogene 1 receptor tyrosine kinase (ROS1), and immune checkpoints; biomarkers associated with resistance to these therapeutics; and approaches to identify predictive biomarkers in anticancer drug development. The identification of predictive biomarkers during anticancer drug development is expected to greatly facilitate such development because it will increase the chance of success or reduce the attrition rate. Additionally, such identification will accelerate the drug approval process by providing effective patient stratification strategies in clinical trials to reduce the sample size required to demonstrate clinical benefits.
Collapse
Affiliation(s)
- Bingliang Fang
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Reza J Mehran
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - John V Heymach
- Department of Thoracic and Head/Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Stephen G Swisher
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
4200
|
Larkin J, Chiarion-Sileni V, Gonzalez R, Grob JJ, Cowey CL, Lao CD, Schadendorf D, Dummer R, Smylie M, Rutkowski P, Ferrucci PF, Hill A, Wagstaff J, Carlino MS, Haanen JB, Maio M, Marquez-Rodas I, McArthur GA, Ascierto PA, Long GV, Callahan MK, Postow MA, Grossmann K, Sznol M, Dreno B, Bastholt L, Yang A, Rollin LM, Horak C, Hodi FS, Wolchok JD. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N Engl J Med 2015; 373:23-34. [PMID: 26027431 PMCID: PMC5698905 DOI: 10.1056/nejmoa1504030] [Citation(s) in RCA: 6149] [Impact Index Per Article: 614.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Nivolumab (a programmed death 1 [PD-1] checkpoint inhibitor) and ipilimumab (a cytotoxic T-lymphocyte-associated antigen 4 [CTLA-4] checkpoint inhibitor) have been shown to have complementary activity in metastatic melanoma. In this randomized, double-blind, phase 3 study, nivolumab alone or nivolumab plus ipilimumab was compared with ipilimumab alone in patients with metastatic melanoma. METHODS We assigned, in a 1:1:1 ratio, 945 previously untreated patients with unresectable stage III or IV melanoma to nivolumab alone, nivolumab plus ipilimumab, or ipilimumab alone. Progression-free survival and overall survival were coprimary end points. Results regarding progression-free survival are presented here. RESULTS The median progression-free survival was 11.5 months (95% confidence interval [CI], 8.9 to 16.7) with nivolumab plus ipilimumab, as compared with 2.9 months (95% CI, 2.8 to 3.4) with ipilimumab (hazard ratio for death or disease progression, 0.42; 99.5% CI, 0.31 to 0.57; P<0.001), and 6.9 months (95% CI, 4.3 to 9.5) with nivolumab (hazard ratio for the comparison with ipilimumab, 0.57; 99.5% CI, 0.43 to 0.76; P<0.001). In patients with tumors positive for the PD-1 ligand (PD-L1), the median progression-free survival was 14.0 months in the nivolumab-plus-ipilimumab group and in the nivolumab group, but in patients with PD-L1-negative tumors, progression-free survival was longer with the combination therapy than with nivolumab alone (11.2 months [95% CI, 8.0 to not reached] vs. 5.3 months [95% CI, 2.8 to 7.1]). Treatment-related adverse events of grade 3 or 4 occurred in 16.3% of the patients in the nivolumab group, 55.0% of those in the nivolumab-plus-ipilimumab group, and 27.3% of those in the ipilimumab group. CONCLUSIONS Among previously untreated patients with metastatic melanoma, nivolumab alone or combined with ipilimumab resulted in significantly longer progression-free survival than ipilimumab alone. In patients with PD-L1-negative tumors, the combination of PD-1 and CTLA-4 blockade was more effective than either agent alone. (Funded by Bristol-Myers Squibb; CheckMate 067 ClinicalTrials.gov number, NCT01844505.).
Collapse
Affiliation(s)
- James Larkin
- Department of Medical Oncology, Royal Marsden Hospital, London, United Kingdom
| | | | - Rene Gonzalez
- Division of Medical Oncology, University of Colorado Denver, Denver, CO
| | - Jean Jacques Grob
- Aix-Marseille University, Hôpital de La Timone APHM, Marseille, France
| | | | - Christopher D. Lao
- Departments of Internal Medicine and Dermatology, University of Michigan, Ann Arbor, MI
| | | | | | | | - Piotr Rutkowski
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | | | - Andrew Hill
- Tasman Oncology Research, Southport Gold Coast, Queensland, Australia
| | - John Wagstaff
- South West Wales Cancer Institute, Singleton Hospital, Swansea, United Kingdom
| | - Matteo S. Carlino
- Westmead and Blacktown Hospitals, Melanoma Institute Australia and the University of Sydney, Sydney, Australia
| | - John B. Haanen
- Division of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | - Ivan Marquez-Rodas
- Servicio de Oncología Médica, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Grant A. McArthur
- Peter MacCallum Cancer Centre and University of Melbourne, Victoria, Australia
| | | | - Georgina V. Long
- Melanoma Institute Australia, the University of Sydney, and the Mater Hospital, Sydney, Australia
| | - Margaret K. Callahan
- Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | - Michael A. Postow
- Memorial Sloan-Kettering Cancer Center and Weill Cornell Medical College, New York, NY
| | | | - Mario Sznol
- Yale Cancer Center, Smilow Cancer Hospital of the Yale-New Haven Hospital, Yale University School of Medicine, New Haven, CT
| | | | - Lars Bastholt
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | | | | | | | | | - Jedd D. Wolchok
- Ludwig Center at Memorial Sloan Kettering Cancer Center and Weill-Cornell Medical College, New York, NY
| |
Collapse
|