5701
|
|
5702
|
Vilgelm AE, Johnson DB, Richmond A. Combinatorial approach to cancer immunotherapy: strength in numbers. J Leukoc Biol 2016; 100:275-90. [PMID: 27256570 PMCID: PMC6608090 DOI: 10.1189/jlb.5ri0116-013rr] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 05/05/2016] [Accepted: 05/11/2016] [Indexed: 12/13/2022] Open
Abstract
Immune-checkpoint blockade therapy with antibodies targeting CTLA-4 and PD-1 has revolutionized melanoma treatment by eliciting responses that can be remarkably durable and is now advancing to other malignancies. However, not all patients respond to immune-checkpoint inhibitors. Extensive preclinical evidence suggests that combining immune-checkpoint inhibitors with other anti-cancer treatments can greatly improve the therapeutic benefit. The first clinical success of the combinatorial approach to cancer immunotherapy was demonstrated using a dual-checkpoint blockade with CTLA-4 and PD-1 inhibitors, which resulted in accelerated FDA approval of this therapeutic regimen. In this review, we discuss the combinations of current and emerging immunotherapeutic agents in clinical and preclinical development and summarize the insights into potential mechanisms of synergistic anti-tumor activity gained from animal studies. These promising combinatorial partners for the immune-checkpoint blockade include therapeutics targeting additional inhibitory receptors of T cells, such as TIM-3, LAG-3, TIGIT, and BTLA, and agonists of T cell costimulatory receptors 4-1BB, OX40, and GITR, as well as agents that promote cancer cell recognition by the immune system, such as tumor vaccines, IDO inhibitors, and agonists of the CD40 receptor of APCs. We also review the therapeutic potential of regimens combining the immune-checkpoint blockade with therapeutic interventions that have been shown to enhance immunogenicity of cancer cells, including oncolytic viruses, RT, epigenetic therapy, and senescence-inducing therapy.
Collapse
Affiliation(s)
- Anna E Vilgelm
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, Tennessee, USA; Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; and
| | - Douglas B Johnson
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ann Richmond
- Tennessee Valley Healthcare System, Department of Veterans Affairs, Nashville, Tennessee, USA; Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA; and
| |
Collapse
|
5703
|
Horn L, Reck M, Spigel DR. The Future of Immunotherapy in the Treatment of Small Cell Lung Cancer. Oncologist 2016; 21:910-21. [PMID: 27354668 PMCID: PMC4978554 DOI: 10.1634/theoncologist.2015-0523] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 04/14/2016] [Indexed: 01/14/2023] Open
Abstract
UNLABELLED : Small cell lung cancer (SCLC), which accounts for 10%-15% of lung cancer cases, is an aggressive disease characterized by rapid growth and early widespread metastasis. Although up to 80% of patients respond to first-line chemotherapy, most eventually relapse, and there are no approved agents beyond the second line. Despite the high incidence of mutations in SCLC, to date no targeted therapy has shown a benefit for this patient population, and systemic treatment has not changed significantly during the past 3 decades. Given that extensive-stage SCLC has a 5-year survival rate of only 1%-2%, novel therapies are desperately needed. Recent evidence shows that the immune system is capable of generating antitumor responses against various tumors, including lung cancer, suggesting that immunotherapy may be a viable therapeutic approach to the treatment of patients with SCLC. Of the immunotherapies being investigated for patients with SCLC, antibodies that target the programmed cell death protein-1 (nivolumab and pembrolizumab) and cytotoxic T-lymphocyte antigen-4 (ipilimumab) immune checkpoint pathways are perhaps the most promising. Because these immune checkpoint pathways, which under normal circumstances function to protect healthy tissues from damage during inflammatory responses and maintain self-tolerance, can help tumor cells evade elimination by the immune system, they represent potential therapeutic targets. This review discusses the rationale for immunotherapy and the early clinical results of immunotherapeutic agents being investigated in SCLC. IMPLICATIONS FOR PRACTICE Small cell lung cancer (SCLC) is an aggressive lung cancer subtype. Despite sensitivity to first-line chemotherapy, SCLC has high recurrence rates, and responses to second-line treatments are poor. Recent evidence shows that the immune system is capable of generating responses against various tumors, including lung cancer, suggesting that immunotherapy may be a viable approach for patients with SCLC. Of several immunotherapies being investigated, antibodies that target the programmed cell death protein-1 (nivolumab and pembrolizumab) and cytotoxic T-lymphocyte antigen-4 (ipilimumab) immune checkpoint pathways are among the most promising for patients with SCLC and are the focus of this review.
Collapse
Affiliation(s)
- Leora Horn
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Martin Reck
- Thoracic Oncology Department, LungenClinic Grosshansdorf, Airway Research Center North, a member of the German Center for Lung Research, Grosshansdorf, Germany
| | - David R Spigel
- Sarah Cannon Research Institute, Nashville, Tennessee, USA
| |
Collapse
|
5704
|
Hanna DL, Lenz HJ. Novel therapeutics in metastatic colorectal cancer: molecular insights and pharmacogenomic implications. Expert Rev Clin Pharmacol 2016; 9:1091-108. [PMID: 27031164 PMCID: PMC7493705 DOI: 10.1586/17512433.2016.1172961] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Although the survival of metastatic colorectal cancer (mCRC) patients has improved five-fold over the last century, CRC remains a significant global health burden. Impressive strides have been made in identifying new regimens, employing maintenance strategies to limit treatment toxicities, and combining multidisciplinary approaches to achieve cure in oligometastatic disease. Attempts at personalized integration of targeted agents have been limited by the ability to identify molecularly enriched patient populations most likely to benefit. In this review, we discuss novel therapeutics and regimens recently approved and in development for mCRC. In addition, we discuss using older agents in novel combination and maintenance strategies, and highlight evidence for implementing pharmacogenomic data and non-invasive monitoring into the personalized management of mCRC patients.
Collapse
Affiliation(s)
- Diana L. Hanna
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Hoag Family Cancer Institute, Newport Beach, CA, USA
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
5705
|
Spranger S. Mechanisms of tumor escape in the context of the T-cell-inflamed and the non-T-cell-inflamed tumor microenvironment. Int Immunol 2016; 28:383-91. [PMID: 26989092 PMCID: PMC4986232 DOI: 10.1093/intimm/dxw014] [Citation(s) in RCA: 213] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 03/14/2016] [Indexed: 12/12/2022] Open
Abstract
Checkpoint blockade therapy has been proven to be highly active across many cancer types but emerging evidence indicates that the therapeutic benefit is limited to a subset of patients in each cancer entity. The presence of CD8(+) T cells within the tumor microenvironment or the invasive margin of the tumor, as well as the up-regulation of PD-L1, have emerged to be the most predictive biomarkers for clinical benefit in response to checkpoint inhibition. Although the up-regulation of immune inhibitory mechanisms is one mechanism of immune escape, commonly used by T-cell-inflamed tumors, exclusion of an anti-tumor specific T-cell infiltrate displays another even more potent mechanism of immune escape. This review will contrast the mechanisms of immunogenic, T-cell-inflamed, and the novel concept of non-immunogenic, non-T-cell-inflamed, adaptive immune escape.
Collapse
Affiliation(s)
- Stefani Spranger
- Department of Pathology, The University of Chicago, GCIS W423H, Chicago, IL 60637, USA
| |
Collapse
|
5706
|
Marrone KA, Ying W, Naidoo J. Immune-Related Adverse Events From Immune Checkpoint Inhibitors. Clin Pharmacol Ther 2016; 100:242-51. [PMID: 27170616 DOI: 10.1002/cpt.394] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/06/2016] [Accepted: 05/07/2016] [Indexed: 01/11/2023]
Abstract
Immunotherapy for cancer treatment has come of age, specifically with the use of immune checkpoint antibodies directed against molecules such as CTLA-4, PD-1, and PD-L1. Single-agent and combinatorial approaches utilizing these agents and other immunotherapies that may enhance antitumor effects are under investigation. With increasing clinical use of these agents, an appreciation for their toxicities comes to the fore. Adverse events that occur as a result of the immunologic effects of these therapies are termed "immune-related adverse events" (irAEs), and range in both frequency and severity in reported single-agent and combination studies. Improvements in our understanding of how and why irAEs develop and how to effectively manage them are needed. Herein we provide a state-of-the-art synopsis of the incidence, clinical features, mechanisms, and management of selected irAEs with immune checkpoint inhibitors currently in use.
Collapse
Affiliation(s)
- K A Marrone
- Department of Oncology, Sidney Kimmel Cancer Center at Johns Hopkins, Baltimore, Maryland, USA
| | - W Ying
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - J Naidoo
- Department of Oncology, Sidney Kimmel Cancer Center at Johns Hopkins, Baltimore, Maryland, USA
| |
Collapse
|
5707
|
Abstract
Immunologic approaches to cancer are over a century old. Over the years, the strategy has been fine-tuned from inciting infections in subjects to inhibiting negative regulatory signals from the innate immune system. Sarcomas are among the first tumors to be considered for immune interventions. From Coley's toxin to cytokine-based therapies to adoptive cell therapy, there have been numerous immunotherapeutic investigations in this patient population. A promising strategy includes adoptive T cell therapy which has been studied in small cohorts of synovial sarcoma, a subtype that is known to widely express the cancer testis antigen, NY-ESO-1. Additionally, recent data in metastatic melanoma and renal cell carcinoma demonstrate the utility and tremendous efficacy of immune checkpoint blockade with increased rates of durable responses compared to standard therapies. Responses in traditionally "non-immunogenic" tumors, such as lung and bladder cancers, provide ample rationale for the study of immune checkpoint inhibitors in sarcoma. While immunotherapy has induced some responses in sarcomas, further research will help clarify optimal patient selection for future clinical trials and new combinatorial immunotherapeutic strategies.
Collapse
|
5708
|
Powles T. Where next in kidney cancer? Ann Oncol 2016; 27:1376-7. [PMID: 27457307 DOI: 10.1093/annonc/mdw247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- T Powles
- Head of Solid Tumor Research, Experimental Cancer Medicine Centre, Barts Cancer Institute, Barts Health NHS Trust and the Royal Free NHS trust, Queen Mary University of London, London, UK
| |
Collapse
|
5709
|
Abstract
Malignant melanoma remains the skin cancer with the highest number of mortalities worldwide. While early diagnosis and complete surgical excision remain the best possibility for curing disease, prognosis at the stage of metastasis is still poor. Recent years have brought about considerable advances in terms of understanding the pathogenesis of melanoma and treating advanced disease. The discovery of activating BRAF mutations in around 50% of tumors has led to the introduction of targeted therapies downregulating BRAF signaling output. These have been further refined as combination therapies, which by targeting multiple targets have further improved the clinical outcome. A comparable, potentially even superior therapeutic alternative has been the introduction of immunotherapeutic approaches, including PD-1 and CTLA-4 checkpoint blockade therapies. Despite all genetic knowledge acquired in recent years, a clearly applicable prognostic signature of clinical value has not been established. General prognostic assessment of cutaneous melanoma remains based on clinical and pathological criteria (most importantly tumor thickness). The main challenges lying ahead are to establish a reliable prognostic test effectively determining which tumors will metastasize. Additionally establishing biomarkers which will allow patients to be stratified according to the most promising systemic therapy (immunotherapies and/or BRAF inhibitor therapies) is of utmost importance for patients with metastasized disease. Identifying serum biomarkers enabling disease to be monitored as well as determining tumor properties (i.e. resistance) would also be of great value. While initial results have proven promising, there remains much work to be done.
Collapse
Affiliation(s)
- Klaus G Griewank
- a Department of Dermatology , University Hospital Essen , Essen , Germany
| |
Collapse
|
5710
|
Sun X, Suo J, Yan J. Immunotherapy in human colorectal cancer: Challenges and prospective. World J Gastroenterol 2016; 22:6362-6372. [PMID: 27605872 PMCID: PMC4968118 DOI: 10.3748/wjg.v22.i28.6362] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 06/02/2016] [Accepted: 06/15/2016] [Indexed: 02/06/2023] Open
Abstract
Human colorectal cancer (CRC) is the third most commonly diagnosed malignancies and the prognosis for patients with recurrent or metastatic disease is extremely poor. Although new chemotherapeutic regimen improves survival rates, therapy with better efficacy and less adverse effects is drastically needed. Immunotherapy has been investigated in human CRC for decades with limited success. However, recent developments of immunotherapy, particularly immune checkpoint inhibitor therapy, have achieved promising clinical benefits in many types of cancer and revived the hope for utilizing such therapy in human CRC. In this review, we will discuss important immunological landscape within the CRC microenvironment and introduce immunoscore system to better describe immunophenotyping in CRC. We will also discuss different immunotherapeutic approaches currently utilized in different phases of clinical trials. Some of those completed or ongoing trials are summarized. Finally, we provide a brief prospective on the future human CRC immunotherapy.
Collapse
|
5711
|
Combined Therapy with Dabrafenib and Trametinib in BRAF-Mutated Metastatic Melanoma in a Real-Life Setting: The INT Milan Experience. TUMORI JOURNAL 2016; 102:501-507. [DOI: 10.5301/tj.5000539] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2016] [Indexed: 11/20/2022]
Abstract
Purpose Combination therapy with dabrafenib and trametinib is safer and more effective than BRAF inhibitor-based monotherapy for metastatic melanoma. Methods We retrospectively analyzed BRAF-mutated metastatic melanoma patients treated at our institution with daily oral dabrafenib 300 mg and trametinib 2 mg from November 2013 to April 2016. This clinical record included both untreated and previously treated stage IV melanomas. Physical examination and laboratory examinations were performed monthly and disease re-evaluations were performed every 3 months. Results A total of 48 patients (24 male, 24 female) with BRAF-mutated metastatic melanoma received dabrafenib and trametinib; median age was 48 years (range 23-75). Median follow-up was 362.5 days (range 72-879). Best overall response rate consisted of 6.2% (3 patients) complete response, 64.6% (31) partial response, and 25% ( 12 ) stable disease; median time to best response was 11 weeks (range 5.7-125.5). Progression of disease was seen in 19 patients (39.6%), with median time to progression (TTP) of 26 weeks (range 8-54). A total of 15 patients (31.2%) died due to progression of disease. Median progression-free survival and median overall survival were not reached. To date, 30 patients (62.5%) are still under treatment. A total of 27 (56.2%) patients had at least one adverse event (AE); grade 3-4 AEs were seen in 4 cases (8.3%). The main toxicities were fever (25%), skin rash (14.6%), arthralgias (10.4%), and aspartate aminotransferase/alanine aminotransferase increase (8.3%). Treatment dose was reduced in 7 subjects (14.6%), with only one case of discontinuation due to AE. Conclusions Our data, using combined targeted therapy, are in line with the scientific literature in terms of both safety and effectiveness in a real-life setting.
Collapse
|
5712
|
Montagnani Marelli M, Marzagalli M, Moretti RM, Beretta G, Casati L, Comitato R, Gravina GL, Festuccia C, Limonta P. Vitamin E δ-tocotrienol triggers endoplasmic reticulum stress-mediated apoptosis in human melanoma cells. Sci Rep 2016; 6:30502. [PMID: 27461002 PMCID: PMC4996065 DOI: 10.1038/srep30502] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 07/06/2016] [Indexed: 12/13/2022] Open
Abstract
Malignant melanoma is the leading cause of death from skin cancer. Drug toxicity and resistance represent a serious challange for melanoma treatments. Evidence demonstrates that natural compounds may play a crucial role in cancer prevention, growth and progression. Vitamin E tocotrienols (TT) were shown to possess antitumor activity. Here, we analyzed the effects of δ-TT on melanoma cell growth and the involvement of the endoplasmic reticulum (ER) stress in this activity. The experiments were performed on human melanoma cell lines, BLM and A375. δ-TT exerted a significant proapoptotic effect on both cell lines, involving the intrinsic apoptosis pathway; importantly, this compound did not affect the viability of normal human melanocytes. In melanoma cells, δ-TT exerted its antitumor effect through activation of the PERK/p-eIF2α/ATF4/CHOP, IRE1α and caspase-4 ER stress-related branches. Salubrinal, an inhibitor of the ER stress, counteracted the cytotoxic activity of δ-TT. In vivo experiments performed in nude mice bearing A375 xenografts evidenced that δ-TT reduces tumor volume and tumor mass; importantly, tumor progression was significantly delayed by δ-TT treatment. In conclusion, δ-TT exerts a proapoptotic activity on melanoma cells, through activation of the ER stress-related pathways. δ-TT might represent an effective option for novel chemopreventive/therapeutic strategies for melanoma.
Collapse
Affiliation(s)
- Marina Montagnani Marelli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, 20133, Italy
| | - Monica Marzagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, 20133, Italy
| | - Roberta M. Moretti
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, 20133, Italy
| | - Giangiacomo Beretta
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milano, 20133, Italy
| | - Lavinia Casati
- Department of Medical Biotechnologies and Translational Medicine, Università degli Studi di Milano, Milano, 20129, Italy
| | - Raffaella Comitato
- Council for Agricultural Research and Economics, Food and Nutrition Research Centre, Roma, 00178, Italy
| | - Giovanni L. Gravina
- Department of Applied and Biotechnological Clinical Sciences, Università degli Studi dell’Aquila, L’Aquila, 67100, Italy
| | - Claudio Festuccia
- Department of Applied and Biotechnological Clinical Sciences, Università degli Studi dell’Aquila, L’Aquila, 67100, Italy
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, 20133, Italy
| |
Collapse
|
5713
|
Schvartsman G, Ferrarotto R, Massarelli E. Checkpoint inhibitors in lung cancer: latest developments and clinical potential. Ther Adv Med Oncol 2016; 8:460-473. [PMID: 27800034 DOI: 10.1177/1758834016661164] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Lung cancer is the leading cause of cancer death in the United States. The vast majority of patients are diagnosed with metastatic disease with a 5-year survival rate of less than 5%. After first-line chemotherapy or biomarker-matched targeted therapy, only suitable for a small group of patients, further systemic therapy options rendered very limited, if any, benefit until recently. Checkpoint inhibitors have significantly improved outcomes in patients with metastatic non-small cell lung cancer (NSCLC) and are currently an established second-line therapeutic option. In this manuscript, we review the mechanism of action of checkpoint inhibitors, present the available data with approved and experimental agents, discuss the progress that has already been made in the field, as well as toxicity awareness, and future perspectives.
Collapse
Affiliation(s)
- Gustavo Schvartsman
- Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Renata Ferrarotto
- Department of Thoracic Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Erminia Massarelli
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, 1500 East Duarte Road, Duarte, CA 91010, USA
| |
Collapse
|
5714
|
Gkountela S, Aceto N. Stem-like features of cancer cells on their way to metastasis. Biol Direct 2016; 11:33. [PMID: 27457474 PMCID: PMC4960876 DOI: 10.1186/s13062-016-0135-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 07/19/2016] [Indexed: 12/18/2022] Open
Abstract
UNLABELLED More than 90 % of cancer-related deaths are due to the development of a systemic metastatic disease. Clearly, much remains to be understood about the biological principles that govern human cancer metastasis, aiming at the ambitious objective to decrease metastasis-related mortality in patients. For many years, research on metastasis has been conducted in great part on experimental mouse models, mainly because of the difficulties in sampling, longitudinal studies, and molecular interrogation of a human metastatic disease. However, recently, extraordinary advances in microfluidic technologies are allowing the isolation and characterization of human circulating tumor cells (CTCs) that escaped a primary tumor mass and are in the process of seeding a distant metastasis. Analysis of human CTCs has now revealed important features of cancer metastasis, such as the high metastatic potential of CTC-clusters compared to single CTCs, the dynamic expression of epithelial and mesenchymal markers on CTCs during treatment, and the possibility to culture CTCs from patients for a real-time and individualized testing of drug susceptibility. Nevertheless, several aspects of CTC biology remain unsolved, such as the characterization of the stem-like cell population among human CTCs. Here, we focus on describing the latest findings in the CTC field, and discuss them in the context of cancer stem cell biology. Defining the molecular features of those few metastasis-initiating, stem-like CTCs holds the exceptional promise to develop metastasis-tailored therapies for patients with cancer. REVIEWERS This article was reviewed by Elisa Cimetta, Luca Pellegrini and Sirio Dupont (nominated by LP).
Collapse
Affiliation(s)
- Sofia Gkountela
- Department of Biomedicine, Cancer Metastasis, University of Basel, Mattenstrasse 28, CH-4058 Basel, Switzerland
| | - Nicola Aceto
- Department of Biomedicine, Cancer Metastasis, University of Basel, Mattenstrasse 28, CH-4058 Basel, Switzerland
| |
Collapse
|
5715
|
Abstract
INTRODUCTION The treatment of melanoma is evolving rapidly over the past few years. Patients with BRAFv600 mutations can be treated with a combination of a BRAF-inhibitor and an MEK-inhibitor. Patients with BRAF wild-type tumors and BRAFv600 mutated tumors can be treated with immunotherapy i.e. check point inhibitors. AREAS COVERED We conducted a comprehensive review of the literature on the efficacy and predictive markers, safety, and pharmacoeconomics of ipilimumab in melanoma Expert commentary: Ipilimumab was the first check point inhibitor reaching the clinic, gaining FDA and EMA approval for metastatic melanoma in 2011. Ipilimumab was also approved by FDA in the adjuvant setting for patients with high risk, stage III melanoma. The anti-PD1 directed antibodies pembrolizumab and nivolumab are superior to single agent ipilimumab, which is no longer considered the standard first line treatment in metastatic melanoma. The addition ipilimumab to nivolumab is associated with a higher response rate and a better PFS, particularly in patients with PD-L1 negative tumors, albeit at the cost of a steep increase in grade 3-4 adverse event rate. Definitive survival data on this combination are pending and the selection of patients potentially requiring the combination and its pharmacoeconomic implications are to be elucidated.
Collapse
Affiliation(s)
- Pol Specenier
- a Oncology , Universitair Ziekenhuis Antwerpen , Edegem , Belgium
| |
Collapse
|
5716
|
Ashizawa T, Iizuka A, Nonomura C, Kondou R, Maeda C, Miyata H, Sugino T, Mitsuya K, Hayashi N, Nakasu Y, Maruyama K, Yamaguchi K, Katano I, Ito M, Akiyama Y. Antitumor Effect of Programmed Death-1 (PD-1) Blockade in Humanized the NOG-MHC Double Knockout Mouse. Clin Cancer Res 2016; 23:149-158. [PMID: 27458246 DOI: 10.1158/1078-0432.ccr-16-0122] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Revised: 06/20/2016] [Accepted: 06/29/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Humanized mouse models using NOD/Shi-scid-IL2rγnull (NOG) and NOD/LtSz-scid IL2rγnull (NSG) mouse are associated with several limitations, such as long incubation time for stem cell engraftment and the development of xenograft versus host disease in mice injected with peripheral blood mononuclear cells (PBMCs). To solve problems, we used humanized major histocompatibility class I- and class II-deficient NOG mice (referred to as NOG-dKO) to evaluate the antitumor effect of anti-programmed death-1 (PD-1) antibody. EXPERIMENTAL DESIGN Humanized NOG-dKO mice, in which human PBMCs and human lymphoma cell line SCC-3, or glioblastoma cell line U87 were transplanted, were used as an immunotherapy model to investigate the effect of anti-PD-1 antibody. A biosimilar anti-PD-1 mAb generated in our laboratory was administered to humanized NOG-dKO mice transplanted with tumors. RESULTS Within 4 weeks after transplantation, human CD45+ cells in antibody-treated mice constituted approximately 70% of spleen cells. The injection of anti-PD-1 antibody reduced by more 50% the size of SCC-3 and U87 tumors. In addition, induction of CTLs against SCC-3 cells and upregulation of natural killer cell activity was observed in the antibody-treated group. Tumor-infiltrating lymphocyte profiling showed that more exhausted marker (PD1+TIM3+LAG3+) positive T cells maintained in anti-PD-1 antibody-treated tumor. A greater number of CD8+ and granzyme-producing T cells infiltrated the tumor in mice treated with the anti-PD-1 antibody. CONCLUSIONS These results suggest that NOG-dKO mice might serve as a good humanized immunotherapy model to evaluate the efficacy of anti-PD-1 antibody prior to the clinical treatment. Clin Cancer Res; 23(1); 149-58. ©2016 AACR.
Collapse
Affiliation(s)
- Tadashi Ashizawa
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka, Japan
| | - Akira Iizuka
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka, Japan
| | - Chizu Nonomura
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka, Japan
| | - Ryota Kondou
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka, Japan
| | - Chie Maeda
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka, Japan
| | - Haruo Miyata
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka, Japan
| | - Takashi Sugino
- Division of Pathology, Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka, Japan
| | - Koichi Mitsuya
- Division of Neurosurgery, Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka, Japan
| | - Nakamasa Hayashi
- Division of Neurosurgery, Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka, Japan
| | - Yoko Nakasu
- Division of Neurosurgery, Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka, Japan
| | - Kouji Maruyama
- Experimental Animal Facility, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka, Japan
| | - Ken Yamaguchi
- Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka, Japan
| | - Ikumi Katano
- Central Institute for Experimental Animals, Kawasaki-ku, Kawasaki, Kanagawa, Japan
| | - Mamoru Ito
- Central Institute for Experimental Animals, Kawasaki-ku, Kawasaki, Kanagawa, Japan
| | - Yasuto Akiyama
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka, Japan. .,Division of Neurosurgery, Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka, Japan
| |
Collapse
|
5717
|
Kageyama SI, Yamaguchi S, Ito S, Suehara Y, Saito T, Akaike K, Miura K, Kato S. A case report of using nivolumab for a malignant melanoma patient with rheumatoid arthritis. Int Cancer Conf J 2016; 5:192-196. [PMID: 31149453 PMCID: PMC6498278 DOI: 10.1007/s13691-016-0256-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/14/2016] [Indexed: 11/03/2022] Open
Abstract
The use of antibodies against programmed cell death 1 (PD-1), which block inhibitory T cell checkpoints, is a promising new therapy for advanced malignant melanoma and NSCLC. However, patients with autoimmune diseases were excluded at the clinical trial using such immune checkpoint inhibitor, because of the possibilities to worsen an adverse event of the autoimmune disease. Thus, the efficacy and toxicity of nivolumab using such cases have not been reported yet. A 70-year-old woman with bone and duodenal metastasis of primary mucosal melanoma with complications of the rheumatoid arthritis was treated with nivolumab. After 4 weeks injection of nivolumab, bone metastasis was diminished. After receiving six courses of nivolumab therapy, she maintained a complete response for 9 months, without rheumatic exacerbation or drug-related adverse events. Establishment of the biomarker of the effect prediction of the PD-1 antibody, the adverse event prediction will be important in future.
Collapse
Affiliation(s)
- Shun-Ichiro Kageyama
- 1Department of Clinical Oncology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421 Japan
| | - Shigeo Yamaguchi
- 1Department of Clinical Oncology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421 Japan
| | - Shin Ito
- 2Department of Otorhinolaryngology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421 Japan
| | - Yoshiyuki Suehara
- 3Department of Orthopedic Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421 Japan
| | - Tsuyoshi Saito
- 4Department of Human Pathology, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421 Japan
| | - Keisuke Akaike
- 3Department of Orthopedic Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421 Japan
| | - Kayo Miura
- 1Department of Clinical Oncology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421 Japan
| | - Shunsuke Kato
- 1Department of Clinical Oncology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421 Japan
| |
Collapse
|
5718
|
Fujimura T, Kambayashi Y, Furudate S, Kakizaki A, Hidaka T, Haga T, Hashimoto A, Morimoto R, Aiba S. Isolated adrenocorticotropic hormone deficiency possibly caused by nivolumab in a metastatic melanoma patient. J Dermatol 2016; 44:e13-e14. [PMID: 27440178 DOI: 10.1111/1346-8138.13532] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Taku Fujimura
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yumi Kambayashi
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Sadanori Furudate
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Aya Kakizaki
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takanori Hidaka
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takahiro Haga
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akira Hashimoto
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryo Morimoto
- Division of Nephrology, Endocrinology and Vascular Medicine, Department of Medicine, Tohoku University Hospital, Sendai, Japan
| | - Setsuya Aiba
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
5719
|
Zhou H, Sharma A. Therapeutic protein-drug interactions: plausible mechanisms and assessment strategies. Expert Opin Drug Metab Toxicol 2016; 12:1323-1331. [PMID: 27391296 DOI: 10.1080/17425255.2016.1211109] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Over the last three decades, therapeutic proteins have played an increasingly important role in pharmacotherapy. Owing to an expected significant increase in the coadministration of biotherapeutics with established pharmacotherapy regimens or even with other biotherapeutic agents, there is an increasing likelihood for the occurrence of clinically relevant drug interactions, so called therapeutic protein-drug interactions (TP-DIs). Areas covered: Our current understanding of TP-DIs and recent collaborations among industry, academia and regulatory agencies are reviewed in this article. Although most of the observed TP-DIs are mediated by disease states, immune status, and/or target physiology, TP-DI assessments are still done empirically. Plausible mechanisms of major TP-DIs involving therapeutic proteins (primarily monoclonal antibodies), either as victims or as perpetrators, are proposed, with mechanism-based strategies and assessment approaches to better evaluate their propensity are recommended. Expert opinion: Our current understanding of the mechanisms of TP-DIs is in its infancy. Much of the basic research needs to be conducted to verify existing TP-DI hypotheses or help predict and manage potential ones, whose efforts are not considered trivial and may be better achieved through close collaborations among scientists from academia, industry, and regulatory agencies.
Collapse
Affiliation(s)
- Honghui Zhou
- a Global Clinical Pharmacology, Quantitative Sciences , Janssen Research and Development, LLC , Spring House , PA , USA
| | - Amarnath Sharma
- a Global Clinical Pharmacology, Quantitative Sciences , Janssen Research and Development, LLC , Spring House , PA , USA
| |
Collapse
|
5720
|
Berghoff AS, Ricken G, Wilhelm D, Rajky O, Widhalm G, Dieckmann K, Birner P, Bartsch R, Preusser M. Tumor infiltrating lymphocytes and PD-L1 expression in brain metastases of small cell lung cancer (SCLC). J Neurooncol 2016; 130:19-29. [DOI: 10.1007/s11060-016-2216-8] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 07/09/2016] [Indexed: 12/31/2022]
|
5721
|
Emens LA, Butterfield LH, Hodi FS, Marincola FM, Kaufman HL. Cancer immunotherapy trials: leading a paradigm shift in drug development. J Immunother Cancer 2016; 4:42. [PMID: 27437105 PMCID: PMC4949883 DOI: 10.1186/s40425-016-0146-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 07/06/2016] [Indexed: 12/14/2022] Open
Affiliation(s)
- Leisha A Emens
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, Room 409, Baltimore, MD 21231-1000 USA
| | - Lisa H Butterfield
- Departments of Medicine, Surgery, and Immunology, Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA USA
| | | | | | - Howard L Kaufman
- Division of Surgical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ USA
| |
Collapse
|
5722
|
Cousin S, Italiano A. Molecular Pathways: Immune Checkpoint Antibodies and their Toxicities. Clin Cancer Res 2016; 22:4550-5. [DOI: 10.1158/1078-0432.ccr-15-2569] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Accepted: 07/06/2016] [Indexed: 11/16/2022]
|
5723
|
Kim JH, Kim BJ, Kim HS, Kim JH. Current Status and Perspective of Immunotherapy in Gastrointestinal Cancers. J Cancer 2016; 7:1599-1604. [PMID: 27698896 PMCID: PMC5039380 DOI: 10.7150/jca.16208] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 07/09/2016] [Indexed: 12/17/2022] Open
Abstract
Cancer immunotherapy is at dawn of the Renaissance after the Medieval Dark Ages. Recent advances of understanding tumor immunology and molecular drug development are leading us to the epoch of cancer immunotherapy. Some types of immunotherapy have shown to provide survival benefit for patients with solid tumors such as malignant melanoma, renal cell carcinoma, or non-small cell lung cancer. Several studies have suggested that immune checkpoint inhibition might be effective in some patients with gastrointestinal cancers. However, the era of cancer immunotherapy in gastrointestinal cancers is still in an inchoate stage. Here we briefly review the current status and perspective of immunotherapeutic approaches in patients with gastrointestinal cancers.
Collapse
Affiliation(s)
- Jung Hoon Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Chuncheon, South Korea
| | - Bum Jun Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Chuncheon, South Korea
| | - Hyeong Su Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Chuncheon, South Korea
| | - Jung Han Kim
- Division of Hemato-Oncology, Department of Internal Medicine, Hallym University Medical Center, Hallym University College of Medicine, Chuncheon, South Korea
| |
Collapse
|
5724
|
Zeltsman M, Mayor M, Jones DR, Adusumilli PS. Surgical immune interventions for solid malignancies. Am J Surg 2016; 212:682-690.e5. [PMID: 27659157 DOI: 10.1016/j.amjsurg.2016.06.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 06/17/2016] [Accepted: 06/17/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND The purpose of this study was to systematically review clinically translatable immunotherapeutic agents that are delivered regionally for solid malignancies. DATA SOURCES PubMed and ClinicalTrials.gov were searched for published and registered clinical trials, respectively. The search yielded 334 relevant publications, of which 116 articles were included for review after exclusion criteria were applied. CONCLUSIONS There has been an increase in the regional administration of cell-based and viral vector-based clinical trials over the last 5 years. Surgical interventions have been developed for intrapleural, intracranial, intraperitoneal, and intratumoral routes of access to enhance the local delivery of these therapies. Multimodality therapies that combine regional immunotherapy with other local and systemic therapies are demonstrating continued growth as the field of immunotherapy continues to expand.
Collapse
Affiliation(s)
- Masha Zeltsman
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, USA
| | - Marissa Mayor
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, USA
| | - David R Jones
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, USA
| | - Prasad S Adusumilli
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, USA; Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY, USA.
| |
Collapse
|
5725
|
Michelle Xu M, Pu Y, Weichselbaum RR, Fu YX. Integrating conventional and antibody-based targeted anticancer treatment into immunotherapy. Oncogene 2016; 36:585-592. [PMID: 27425593 PMCID: PMC5243926 DOI: 10.1038/onc.2016.231] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 04/17/2016] [Accepted: 04/28/2016] [Indexed: 12/18/2022]
Abstract
In advanced cancer, current conventional therapies or immunotherapies cannot eradicate all tumor cells for most patients. Integration of these two treatments for synergistic effects could eradicate more tumor cells and increase overall survival rates. But proper integration is a challenge, partly due to a poor understanding of the impact of conventional treatment on immune responses. Intensive chemo/radiotherapy may impair ongoing immune responses whilst lower intensity of therapy might not kill enough tumor cells, both leading to tumor relapse. Current understanding of mechanisms of resistance to conventional and targeted cancer therapies has focused on cell intrinsic pathways that trigger DNA damage/repair or signaling pathways related to cell growth. Recent reports show that host T cells properly primed against tumor specific antigens after conventional treatment can integrate with direct cytotoxic effects induced by radiation or chemotherapy to profoundly control tumors. Following cytotoxic anticancer treatment, tumor derived DAMPs (damage-associated molecular patterns) can be sensed by innate cells which drives type I interferon (IFNs production) for cross priming of CD8+ T cells. Some types and protocols of chemotherapy or radiation can increase tumor infiltrating lymphocytes that overcome resistance to immunotherapy. As such, a deeper understanding to the immune mechanisms of conventional and targeted cancer therapies will lead toward novel combinatorial anticancer strategies with improved clinical benefit.
Collapse
Affiliation(s)
- M Michelle Xu
- Department of Pathology and Radiation Oncology, University of Chicago, Chicago, IL, USA.,Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA
| | - Y Pu
- Department of Pathology and Immunology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - R R Weichselbaum
- Department of Pathology and Radiation Oncology, University of Chicago, Chicago, IL, USA
| | - Y-X Fu
- Department of Pathology and Radiation Oncology, University of Chicago, Chicago, IL, USA.,Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL, USA.,Department of Pathology and Immunology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
5726
|
Jensen IS, Zacherle E, Blanchette CM, Zhang J, Yin W. Evaluating cost benefits of combination therapies for advanced melanoma. Drugs Context 2016; 5:212297. [PMID: 27540409 PMCID: PMC4974051 DOI: 10.7573/dic.212297] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Although a number of monoimmunotherapies and targeted therapies are available to treat BRAF+ advanced melanoma, response rates remain relatively low in the range of 22-53% with progression-free survival (PFS) in the range of 4.8-8.8 months. Recently, combination targeted therapies have improved response rates to about 66-69%, PFS to 11.0-12.6 months and overall survival (OS) to 25.1-25.6 months. While combination immunotherapies have improved response rates of 67 compared with 19-29% with monotherapies and improved PFS of 11.7 compared with 4.4-5.8 months with monotherapies, the OS benefit is yet to be established in phase 3 trials. As healthcare costs continue to rise, US payers have a predominant interest in assessing the value of available treatments. Therefore, a cost-benefit model was developed to evaluate the value of treating BRAF+ advanced melanoma with two combination therapies: nivolumab + ipilimumab (N+I) and dabrafenib + trametinib (D+T). SCOPE The model was used to estimate total costs, total costs by expenditure category, cost per month of PFS and cost per responder for the payer, and societal perspectives of treating advanced melanoma patients with the BRAF V600 mutation using combination targeted therapy (D+T) or combination immunotherapy (N+I). The model followed patients from initiation of treatment to the point of progression or death. Deterministic and probabilistic sensitivity analyses were conducted to evaluate the robustness of the results and to understand the dispersion of simulated results. FINDINGS Based on a hypothetical payer with one million covered lives, it was expected that fourteen metastatic melanoma patients with the BRAF V600 mutation would be treated each year. Cost-benefit with N+I and D+T was simulated from the payer perspective. The cost per month of PFS for N+I was $22,162, while that for D+T was $17,716 (-$4,446 cost difference); the cost per responder for N+I was $388,746 and that for D+T was $282,429 (-$106,316 cost difference). The cost per month of PFS and per responder from the societal perspective resembled the patterns observed from the payer's perspective: the cost per month of PFS for N+I was $22,843, while that for D+T was $18,283 (-$4,560 cost difference). The cost per responder for N+I was $400,695 and that for D+T was $291,473 (-$109,222 cost difference). The totals of travel and treatment time for N+I and D+T were 58 hours and 3.9 hours per patient, respectively, of which total infusion time for N+I accounted for a majority - 59% - of the 58 hours. Sensitivity analyses indicated that results were most sensitive to model inputs for median PFS, body weight, and drug cost. Moreover, D+T is likely associated with a lower cost per month of PFS and cost per responder than N+I, except at low body weights (less than 57 kg). CONCLUSION The model presented in this study was used to analyze the clinical and economic benefit of using combination therapies in advanced melanoma patients with the BRAF V600 mutation. This analysis suggests D+T therapy is associated with less patient time and lower costs relative to N+I to gain similar PFS and overall response rate (ORR) benefits.
Collapse
Affiliation(s)
- Ivar S. Jensen
- Precision Health Economics, 101 Tremont St. Suite 400, Boston, MA 02108, USA
| | - Emily Zacherle
- Precision Health Economics, 209 Delburg St. Suite 106, Davidson, NC 28036, USA
- University of North Carolina at Charlotte, 9201 University City Blvd,Charlotte, NC 28223, USA
| | - Christopher M. Blanchette
- Precision Health Economics, 209 Delburg St. Suite 106, Davidson, NC 28036, USA
- University of North Carolina at Charlotte, 9201 University City Blvd,Charlotte, NC 28223, USA
| | - Jie Zhang
- Novartis Pharmaceuticals Corporation, One Health Plaza, BLDG 315, 5550B, East Hanover, NJ 07936, USA
| | - Wes Yin
- University of California at Los Angelos, 11100 Santa Monica Blvd. Suite 500, Los Angelos, CA 90025, USA
| |
Collapse
|
5727
|
Suozzi KC, Stahl M, Ko CJ, Chiang A, Gettinger SN, Siegel MD, Bunick CG. Immune-related sarcoidosis observed in combination ipilimumab and nivolumab therapy. JAAD Case Rep 2016; 2:264-8. [PMID: 27486590 PMCID: PMC4949498 DOI: 10.1016/j.jdcr.2016.05.002] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Kathleen C Suozzi
- Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut
| | - Maximilian Stahl
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Christine J Ko
- Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut
| | - Anne Chiang
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut; Smilow Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Scott N Gettinger
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut; Smilow Cancer Center, Yale University School of Medicine, New Haven, Connecticut
| | - Mark D Siegel
- Department of Medicine, Yale University School of Medicine, New Haven, Connecticut
| | - Christopher G Bunick
- Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut
| |
Collapse
|
5728
|
van Zeijl MCT, van den Eertwegh AJ, Haanen JB, Wouters MWJM. (Neo)adjuvant systemic therapy for melanoma. Eur J Surg Oncol 2016; 43:534-543. [PMID: 27453302 DOI: 10.1016/j.ejso.2016.07.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 06/28/2016] [Accepted: 07/04/2016] [Indexed: 11/28/2022] Open
Abstract
Surgery still is the cornerstone of treatment for patients with stage II and III melanoma, but despite great efforts to gain or preserve locoregional control with excision of the primary tumour, satellites, intransits, sentinel node biopsy and lymphadenectomy, surgery alone does not seem to improve survival any further. Prognosis for patients with high risk melanoma remains poor with 5-year survival rates of 40 to 80%. Only interferon-2b has been approved as adjuvant therapy since 1995, but clinical integration is low considering the high risk-benefit ratio. In recent years systemic targeted- and immunotherapy have proven to be beneficial in advanced melanoma and could be a promising strategy for (neo)adjuvant treatment of patients with resectable high risk melanomas as well. Randomised, placebo- controlled phase III trials on adjuvant systemic targeted- and immunotherapy are currently being performed using new agents like ipilimumab, pembrolizumab, nivolumab, vemurafenib and dabrafenib plus trametinib. In this article we review the literature on currently known adjuvant therapies and currently ongoing trials of (neo)adjuvant therapies in high risk melanomas.
Collapse
Affiliation(s)
- M C T van Zeijl
- Dutch Institute for Clinical Auditing, Rijnsburgerweg 10, 2333AA Leiden, The Netherlands; Department of Surgery, Leiden University Medical Centre, Albinusdreef 2, 2333ZA Leiden, The Netherlands.
| | - A J van den Eertwegh
- Department of Medical Oncology, VU University Medical Center, De Boelelaan 1118, 1081HZ Amsterdam, The Netherlands
| | - J B Haanen
- Department of Medical Oncology and Immunology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| | - M W J M Wouters
- Dutch Institute for Clinical Auditing, Rijnsburgerweg 10, 2333AA Leiden, The Netherlands; Department of Surgical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066CX Amsterdam, The Netherlands
| |
Collapse
|
5729
|
Roldan-Deamicis A, Alonso E, Brie B, Braico DA, Balogh GA. Maitake Pro4X has anti-cancer activity and prevents oncogenesis in BALBc mice. Cancer Med 2016; 5:2427-41. [PMID: 27401257 PMCID: PMC5055164 DOI: 10.1002/cam4.744] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 02/17/2016] [Accepted: 03/08/2016] [Indexed: 12/26/2022] Open
Abstract
The understanding of the molecular mechanisms of the immune tolerance induced by the tumoral microenvironment is fundamental to prevent cancer development or to treat cancer patients using immunotherapy. Actually, there are investigations about "addressed-drugs" against cancer cells without affecting normal cells. It could be ideal to find selective and specific compounds that only recognize and destroy tumor cells without damaging the host normal cells. For thousands of years, mushrooms have been used for medicinal purposes because of their curative properties. D-Fraction, an extract of Maitake (from the edible Grifola frondosa mushroom), rich in β-glucans, exert notable effects in the immune system. Until now, some published articles suggest that Maitake D-Fraction could have anti-tumoral activity, prevent oncogenesis and metastasis in some tumor types. However, there are no clear data about Maitake D-Fraction action on breast cancer prevention and its exact molecular mechanisms are not yet elucidated. The experiments were performed employing 25 female BALBc mice that were treated with and without Maitake D-Fraction Pro4X or Maitake Standard for 15 days by daily intraperitoneal injection. After treatment period, all mice were implanted with murine tumor cells LM3 to induce mammary tumorigenesis. Animals were checked weekly and killed after 46 days of LM3 transplant; percentage of cancer prevention, rate of tumor growing, and overall survival were determined. Under dissection, the internal organs were evaluated histologically and genetically by RT-PCR. We found that 5 mg/kg per day of Maitake D-Fraction Pro4X, administered dairy during 15 days to BALBc mice was able to block more than 60% breast cancer development. However, Maitake Standard prevents oncogenesis in 26% to respect control. In this work, we found that Maitake D-Fraction Pro4X, administered to BALBc mice, prevents breast carcinogenesis, block tumor invasiveness, reduce angiogenesis, and increase overall survival.
Collapse
Affiliation(s)
- Agustina Roldan-Deamicis
- Instituto de Investigaciones Biomédicas, Facultad de Ciencias Medicas, Pontificia Universidad Católica Argentina - UCA - CONICET, Buenos Aires, Argentina
| | - Eliana Alonso
- Laboratorio de Hongos Comestibles y Medicinales, Centro Científico Tecnológico, CERZOS-CONICET, Camino La Carrindanga Km7, Bahía Blanca-8000, Buenos Aires, Argentina
| | - Belén Brie
- Instituto de Investigaciones Biomédicas, Facultad de Ciencias Medicas, Pontificia Universidad Católica Argentina - UCA - CONICET, Buenos Aires, Argentina
| | - Diego Aguilera Braico
- Instituto de Investigaciones Biomédicas, Facultad de Ciencias Medicas, Pontificia Universidad Católica Argentina - UCA - CONICET, Buenos Aires, Argentina
| | - Gabriela Andrea Balogh
- Instituto de Investigaciones Biomédicas, Facultad de Ciencias Medicas, Pontificia Universidad Católica Argentina - UCA - CONICET, Buenos Aires, Argentina.
| |
Collapse
|
5730
|
Moriguchi M, Umemura A, Itoh Y. Current status and future prospects of chemotherapy for advanced hepatocellular carcinoma. Clin J Gastroenterol 2016; 9:184-90. [PMID: 27401471 DOI: 10.1007/s12328-016-0670-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 06/09/2016] [Indexed: 02/06/2023]
Abstract
Sorafenib is the only drug that demonstrates a survival benefit for advanced hepatocellular carcinoma (HCC). However, the therapeutic effect of sorafenib is limited, so development of a more effective treatment method and second-line treatments is needed. Since the advent of sorafenib, clinical studies have been conducted with a variety of drugs and treatment methods, mainly with molecular targeted therapy, but almost all trials have ended in failure. The reasons for the difficulty in the development of a novel drug or treatment method include the diversity of mechanisms in the carcinogenesis and development of HCC, as well as the presence of background liver diseases such as chronic hepatitis and cirrhosis. Trials with immune-checkpoint inhibitors, which have an entirely different anti-tumor mechanism from that of molecular targeted drugs or cytotoxic drugs, have recently begun. Based on the results to date, clinical trials are now being conducted with enriched target subjects. In the future, providing more individualized treatment approaches for patients with advanced HCC will be essential.
Collapse
Affiliation(s)
- Michihisa Moriguchi
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi-Hirokouji, Kamigyo-ku, Kyoto, 602-8566, Japan.
| | - Atsushi Umemura
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi-Hirokouji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Yoshito Itoh
- Department of Molecular Gastroenterology and Hepatology, Kyoto Prefectural University of Medicine, Graduate School of Medical Science, 465 Kajii-cho, Kawaramachi-Hirokouji, Kamigyo-ku, Kyoto, 602-8566, Japan
| |
Collapse
|
5731
|
Migali C, Milano M, Trapani D, Criscitiello C, Esposito A, Locatelli M, Minchella I, Curigliano G. Strategies to modulate the immune system in breast cancer: checkpoint inhibitors and beyond. Ther Adv Med Oncol 2016; 8:360-74. [PMID: 27583028 DOI: 10.1177/1758834016658423] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Is breast cancer (BC) immunogenic? Many data suggest that it is. Many observations demonstrated the prognostic role of tumor-infiltrating lymphocytes (TILs) in triple negative (TN) and human epidermal growth factor receptor 2 (HER-2)-positive BC. TNBCs are poorly differentiated tumors with high genetic instability and very high heterogeneity. This heterogeneity enhances the 'danger signals' and select clone variants that could be more antigenic or, in other words, that could more strongly stimulate a host immune antitumor response. The response to chemotherapy is at least partly dependent on an immunological reaction against those tumor cells that are dying during the chemotherapy. One of the mechanisms whereby chemotherapy can stimulate the immune system to recognize and destroy malignant cells is commonly known as immunogenic cell death (ICD). ICD elicits an adaptive immune response. Which are the clinical implications of all 'immunome' data produced in the last years? First, validate prognostic or predictive role of TILs. Second, validate immune genomic signatures that may be predictive and prognostic in patients with TN disease. Third, incorporate an 'immunoscore' into traditional classification of BC, thus providing an essential prognostic and potentially predictive tool in the pathology report. Fourth, implement clinical trials for BC in the metastatic setting with drugs that target immune-cell-intrinsic checkpoints. Blockade of one of these checkpoints, cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) or the programmed cell death 1 (PD-1) receptor may provide proof of concepts for the activity of an immune-modulation approach in the treatment of a BC.
Collapse
Affiliation(s)
- Cristina Migali
- Division of Experimental Therapeutics, Istituto Europeo di Oncologia, Milano, Italy
| | - Monica Milano
- Division of Experimental Therapeutics, Istituto Europeo di Oncologia, Milano, Italy
| | - Dario Trapani
- Division of Experimental Therapeutics, Istituto Europeo di Oncologia, Milano, Italy
| | - Carmen Criscitiello
- Division of Experimental Therapeutics, Istituto Europeo di Oncologia, Milano, Italy
| | - Angela Esposito
- Division of Experimental Therapeutics, Istituto Europeo di Oncologia, Milano, Italy
| | - Marzia Locatelli
- Division of Experimental Therapeutics, Istituto Europeo di Oncologia, Milano, Italy
| | - Ida Minchella
- Division of Experimental Therapeutics, Istituto Europeo di Oncologia, Milano, Italy
| | - Giuseppe Curigliano
- Division of Experimental Therapeutics, Istituto Europeo di Oncologia, Via Ripamonti 435, 20141 Milano, Italy
| |
Collapse
|
5732
|
Smalley KSM. Increased immunity and BRAF inhibition: Yet another argument for combination therapy? Pharmacol Res 2016; 113:719-720. [PMID: 27392741 DOI: 10.1016/j.phrs.2016.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 06/14/2016] [Indexed: 11/19/2022]
Affiliation(s)
- Keiran S M Smalley
- The Departments of Tumor Biology and Cutaneous Oncology, The Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, United States.
| |
Collapse
|
5733
|
Recent developments and future challenges in immune checkpoint inhibitory cancer treatment. Curr Opin Oncol 2016; 27:482-8. [PMID: 26352539 DOI: 10.1097/cco.0000000000000221] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE OF REVIEW In this review, we focus on the recent findings and future challenges in cancer treatment with immune checkpoint inhibitors. RECENT FINDINGS Major progress has been made in recent years as the first immune checkpoint inhibitors are approved by the US Food and Drug Administration for the treatment of cancer patients. Anticytotoxic T-lymphocyte-associated protein 4 and antiprogrammed death protein 1/programmed death-ligand 1 (PD-L1) monoclonal antibodies are being extensively studied in many different tumor types, often showing impressive response rates, but also a typical serious toxicity profile in the form of auto-immunity. Unfortunately, it is not yet possible to prevent or predict these immune-related adverse events. Studies on mutational load, neo-epitopes, lactate dehydrogenase, PD-L1 expression, and T-cell infiltration suggest that these markers are correlating with efficacy, but have not yet reached the status of a validated biomarker for checkpoint inhibitors. Other immune checkpoints are being investigated and new checkpoint inhibitors are on the brink of being evaluated in clinical trials. SUMMARY The main challenge for the near future will be to predict efficacy of immune checkpoint blockade and to predict and prevent immune-related adverse events. More research should be done in order to find potential biomarkers that predict treatment response and/or toxicity; the optimal administration route, dosage, and frequency; and possible combinations of therapies that have an added or synergetic effect.
Collapse
|
5734
|
Weber JS, Postow M, Lao CD, Schadendorf D. Management of Adverse Events Following Treatment With Anti-Programmed Death-1 Agents. Oncologist 2016; 21:1230-1240. [PMID: 27401894 DOI: 10.1634/theoncologist.2016-0055] [Citation(s) in RCA: 182] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 05/02/2016] [Indexed: 12/28/2022] Open
Abstract
: Immune checkpoint inhibitors have emerged as a mainstay of melanoma therapy and are playing an increasingly important role in the treatment of other tumor types. The clinical benefit afforded by these treatments can be accompanied by a unique spectrum of adverse events, called immune-related adverse events (irAEs), which reflect the drug's immune-based mechanism of action. IrAEs typically originate in the skin, gastrointestinal tract, liver, and endocrine system, although other organ systems may also be affected. This article provides an overview of irAEs associated with anti-programmed death-1 (anti-PD-1) antibodies (nivolumab and pembrolizumab) as monotherapy or in combination with anti-cytotoxic T-lymphocyte antigen-4 inhibition (ipilimumab), followed by a discussion of irAEs of special clinical interest based on the potential for morbidity, frequent steroid use, and inpatient admission. We review clinical trial data and provide recommendations on how to manage irAEs associated with anti-PD-1 agents based on clinical experience and established management guidelines. We further illustrate the practical considerations of managing irAEs by presenting three cases of immune-related toxicity in melanoma patients treated with nivolumab or pembrolizumab. A better understanding of the identification and management of irAEs will help inform health care providers about the risks associated with anti-PD-1 treatment, to ensure the safe and appropriate use of these important new treatments. IMPLICATIONS FOR PRACTICE Immune checkpoint inhibitors have demonstrated significant clinical benefit in advanced melanoma and other tumor types. These treatments are associated with immune-related adverse events (irAEs), which most commonly affect the skin and gastrointestinal tract, and, to a lesser extent, the liver, endocrine system, and other organs. This review focuses on the management of irAEs after treatment with anti-programmed death-1 (anti-PD-1) antibodies (nivolumab or pembrolizumab) as monotherapy or in combination with anti-cytotoxic T-lymphocyte antigen-4 inhibition (ipilimumab) in patients with advanced melanoma. A better understanding of the management of irAEs will help ensure the safe and appropriate use of anti-PD-1 agents in melanoma and other tumor types.
Collapse
Affiliation(s)
- Jeffrey S Weber
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York, USA
| | - Michael Postow
- Memorial Sloan Kettering Cancer Center, New York, New York, USA Weill Cornell Medical College, New York, New York, USA
| | - Christopher D Lao
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | |
Collapse
|
5735
|
Borch TH, Engell-Noerregaard L, Zeeberg Iversen T, Ellebaek E, Met Ö, Hansen M, Andersen MH, Thor Straten P, Svane IM. mRNA-transfected dendritic cell vaccine in combination with metronomic cyclophosphamide as treatment for patients with advanced malignant melanoma. Oncoimmunology 2016; 5:e1207842. [PMID: 27757300 DOI: 10.1080/2162402x.2016.1207842] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/13/2016] [Accepted: 06/25/2016] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION Vaccination with dendritic cells (DCs) has generally not fulfilled its promise in cancer immunotherapy due to ineffective translation of immune responses into clinical responses. A proposed reason for this is intrinsic immune regulatory mechanisms, such as regulatory T cells (Tregs). A metronomic regimen of cyclophosphamide (mCy) has been shown to selectively deplete Tregs. To test this in a clinical setting, we conducted a phase I trial to evaluate the feasibility and safety of vaccination with DCs transfected with mRNA in combination with mCy in patients with metastatic malignant melanoma (MM). In addition, clinical and immunological effect of the treatment was evaluated. EXPERIMENTAL DESIGN Twenty-two patients were enrolled and treated with six cycles of cyclophosphamide 50 mg orally bi-daily for a week every second week (day 1-7). During the six cycles patients received at least 5 × 106 autologous DCs administered by intradermal (i.d.) injection in the week without chemotherapy. Patients were evaluated 12 and 27 weeks and every 3rd mo thereafter with CT scans according to RECIST 1.0. Blood samples for immune monitoring were collected at baseline, at the time of 4th and 6th vaccines. Immune monitoring consisted of IFNγ ELISpot assay, proliferation assay, and flow cytometry for enumeration of immune cell subsets. RESULTS Toxicity was manageable. Eighteen patients were evaluable after six cycles. Of these, nine patients had progressive disease as best response and nine patients achieved stable disease. In three patients minor tumor regression was observed. By IFNγ ELISpot and proliferation assay immune responses were seen in 6/17 and 4/17 patients, respectively; however, no correlation with clinical response was found. The percentage of Tregs was unchanged during treatment. CONCLUSION Treatment with autologous DCs transfected with mRNA in combination with mCy was feasible and safe. Importantly, mCy did not alter the percentage of Tregs in our patient cohort. There was an indication of clinical benefit; however, more knowledge is needed in order for DCs to be exploited as a therapeutic option.
Collapse
Affiliation(s)
- Troels Holz Borch
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Denmark; Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Lotte Engell-Noerregaard
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Denmark; Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Trine Zeeberg Iversen
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Denmark; Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Eva Ellebaek
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Denmark; Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Özcan Met
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Denmark; Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| | - Morten Hansen
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital , Herlev, Denmark
| | - Mads Hald Andersen
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital , Herlev, Denmark
| | - Per Thor Straten
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital , Herlev, Denmark
| | - Inge Marie Svane
- Center for Cancer Immune Therapy (CCIT), Department of Hematology, Copenhagen University Hospital, Herlev, Denmark; Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
| |
Collapse
|
5736
|
Santabarbara G, Maione P, Rossi A, Palazzolo G, Gridelli C. The role of pembrolizumab in the treatment of advanced non-small cell lung cancer. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:215. [PMID: 27386489 DOI: 10.21037/atm.2016.05.64] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Lung cancer is the leading cause of death cancer related worldwide. The standard therapies have unmet medical needs both due to the limited activity and relevant toxicity of platinum-based chemotherapy and to the low frequency of specific alterations required to use targeted therapies. Immune checkpoint inhibition due to restoring the immune system's capacity to eradicate tumors is undergoing in extensive investigation in non-small cell lung cancer (NSCLC) as a new treatment approach. Programmed cell death protein-1 (PD-1) and its ligand, programmed cell death-ligand 1 (PD-L1) have recently led to significantly and durable improvements in the clinical outcome of several kind of tumors including lung cancer. Pembrolizumab, approved by the U.S. FDA for the treatment of advanced NSCLC progressed after other therapies and with expression of PD-L1, has demonstrated durable response and prolonged overall survival (OS) especially in patients with high PD-L1 expression. Further investigation are needed to improve treatment outcomes through combination of immunotherapy or combined with other targeted therapies.
Collapse
Affiliation(s)
- Giuseppe Santabarbara
- 1 Division of Medical Oncology, "S. G. Moscati" Hospital, Contrada Amoretta 83100, Avellino, Italy ; 2 Division of Medical Oncology, "ULSS 15 Cittadella", via Casa di ricovero, 40 35013 Cittadella, Padova, Italy
| | - Paolo Maione
- 1 Division of Medical Oncology, "S. G. Moscati" Hospital, Contrada Amoretta 83100, Avellino, Italy ; 2 Division of Medical Oncology, "ULSS 15 Cittadella", via Casa di ricovero, 40 35013 Cittadella, Padova, Italy
| | - Antonio Rossi
- 1 Division of Medical Oncology, "S. G. Moscati" Hospital, Contrada Amoretta 83100, Avellino, Italy ; 2 Division of Medical Oncology, "ULSS 15 Cittadella", via Casa di ricovero, 40 35013 Cittadella, Padova, Italy
| | - Giovanni Palazzolo
- 1 Division of Medical Oncology, "S. G. Moscati" Hospital, Contrada Amoretta 83100, Avellino, Italy ; 2 Division of Medical Oncology, "ULSS 15 Cittadella", via Casa di ricovero, 40 35013 Cittadella, Padova, Italy
| | - Cesare Gridelli
- 1 Division of Medical Oncology, "S. G. Moscati" Hospital, Contrada Amoretta 83100, Avellino, Italy ; 2 Division of Medical Oncology, "ULSS 15 Cittadella", via Casa di ricovero, 40 35013 Cittadella, Padova, Italy
| |
Collapse
|
5737
|
Polireddy K, Chen Q. Cancer of the Pancreas: Molecular Pathways and Current Advancement in Treatment. J Cancer 2016; 7:1497-514. [PMID: 27471566 PMCID: PMC4964134 DOI: 10.7150/jca.14922] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 04/26/2016] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is one of the most lethal cancers among all malignances, with a median overall survival of <1 year and a 5-year survival of ~5%. The dismal survival rate and prognosis are likely due to lack of early diagnosis, fulminant disease course, high metastasis rate, and disappointing treatment outcome. Pancreatic cancers harbor a variety of genetic alternations that render it difficult to treat even with targeted therapy. Recent studies revealed that pancreatic cancers are highly enriched with a cancer stem cell (CSC) population, which is resistant to chemotherapeutic drugs, and therefore escapes chemotherapy and promotes tumor recurrence. Cancer cell epithelial to mesenchymal transition (EMT) is highly associated with metastasis, generation of CSCs, and treatment resistance in pancreatic cancer. Reviewed here are the molecular biology of pancreatic cancer, the major signaling pathways regulating pancreatic cancer EMT and CSCs, and the advancement in current clinical and experimental treatments for pancreatic cancer.
Collapse
Affiliation(s)
- Kishore Polireddy
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, USA 66160
| | - Qi Chen
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS, USA 66160
| |
Collapse
|
5738
|
Silvestri I, Cattarino S, Giantulli S, Nazzari C, Collalti G, Sciarra A. A Perspective of Immunotherapy for Prostate Cancer. Cancers (Basel) 2016; 8:cancers8070064. [PMID: 27399780 PMCID: PMC4963806 DOI: 10.3390/cancers8070064] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/27/2016] [Accepted: 07/01/2016] [Indexed: 12/24/2022] Open
Abstract
In cancer patients, the immune system is often altered with an excess of inhibitory factors, such as immunosuppressive cytokines, produced by regulatory T cells (Treg) or myeloid-derived suppressor cells (MDSC). The manipulation of the immune system has emerged as one of new promising therapies for cancer treatment, and also represents an attractive strategy to control prostate cancer (PCa). Therapeutic cancer vaccines and immune checkpoint inhibitors have been the most investigated in clinical trials. Many trials are ongoing to define the effects of immune therapy with established treatments: androgen deprivation therapy (ADT) and chemotherapy (CT) or radiotherapy (RT). This article discusses some of these approaches in the context of future treatments for PCa.
Collapse
Affiliation(s)
- Ida Silvestri
- Department of Molecular Medicine, Sapienza University of Rome, Rome 00161, Italy.
| | - Susanna Cattarino
- Department of Urology, Sapienza University of Rome, Rome 00161, Italy.
| | - Sabrina Giantulli
- Department of Molecular Medicine, Sapienza University of Rome, Rome 00161, Italy.
| | - Cristina Nazzari
- Department of Public Health hand Infectious Diseases, "Sapienza" University of Rome, Rome 00185, Italy.
| | - Giulia Collalti
- Medicine of Systems, Rheumatology, Allergology and Clinical Immunology, Translational Medicine of the University Tor Vergata, Rome 00133, Italy.
| | | |
Collapse
|
5739
|
Hendriks D, He Y, Koopmans I, Wiersma VR, van Ginkel RJ, Samplonius DF, Helfrich W, Bremer E. Programmed Death Ligand 1 (PD-L1)-targeted TRAIL combines PD-L1-mediated checkpoint inhibition with TRAIL-mediated apoptosis induction. Oncoimmunology 2016; 5:e1202390. [PMID: 27622071 PMCID: PMC5007955 DOI: 10.1080/2162402x.2016.1202390] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 06/07/2016] [Accepted: 06/12/2016] [Indexed: 01/18/2023] Open
Abstract
Antibodies that block PD-L1/PD-1 immune checkpoints restore the activity of functionally-impaired antitumor T cells. These antibodies show unprecedented clinical benefit in various advanced cancers, particularly in melanoma. However, only a subset of cancer patients responds to current PD-L1/PD-1-blocking strategies, highlighting the need for further advancements in PD-L1/PD-1-based immunotherapy. Here, we report on a novel approach designed to combine PD-L1 checkpoint inhibition with the tumor-selective induction of apoptosis by TNF-related Apoptosis Inducing Ligand (TRAIL). In brief, a new bi-functional fusion protein, designated anti-PD-L1:TRAIL, was constructed comprising a PD-L1-blocking antibody fragment genetically fused to the extracellular domain of the pro-apoptotic tumoricidal protein TRAIL. Treatment of PD-L1-expressing cancer cells with anti-PD-L1:TRAIL induced PD-L1-directed TRAIL-mediated cancer cell death. Treatment of T cells with anti-PD-L1:TRAIL augmented T cell activation, as evidenced by increased proliferation, secretion of IFNγ and enhanced killing of cancer cell lines and primary patient-derived cancer cells in mixed T cell/cancer cell culture experiments. Of note, elevated levels of IFNγ further upregulated PD-L1 on cancer cells and simultaneously sensitized cancer cells to TRAIL-mediated apoptosis by anti-PD-L1:TRAIL. Additionally, anti-PD-L1:TRAIL converted immunosuppressive PD-L1-expressing myeloid cells into pro-apoptotic effector cells that triggered TRAIL-mediated cancer cell death. In conclusion, combining PD-L1 checkpoint inhibition with TRAIL-mediated induction of apoptosis using anti-PD-L1:TRAIL yields promising multi-fold and mutually reinforcing anticancer activity that may be exploited to enhance the efficacy of therapeutic PD-L1/PD-1 checkpoint inhibition.
Collapse
Affiliation(s)
- Djoke Hendriks
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Yuan He
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Iris Koopmans
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Valerie R Wiersma
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Robert J van Ginkel
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Douwe F Samplonius
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Wijnand Helfrich
- University of Groningen, University Medical Center Groningen (UMCG), Department of Surgery, Laboratory for Translational Surgical Oncology , Groningen, the Netherlands
| | - Edwin Bremer
- University of Exeter Medical School, St Luke's Campus, Exeter, Devon, UK; University of Groningen, University Medical Center Groningen, Department of Experimental Hematology, Section Immunohematology, Cancer Research Center Groningen (CRCG), Groningen, the Netherlands
| |
Collapse
|
5740
|
Wilden SM, Lang BM, Mohr P, Grabbe S. Checkpoint-Inhibitoren in der Immuntherapie: Ein Meilenstein in der Behandlung des malignen Melanoms. J Dtsch Dermatol Ges 2016; 14:685-97. [PMID: 27373243 DOI: 10.1111/ddg.13012_g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Seit Jahrzehnten ist bekannt, dass Tumoren vom Immunsystem erkannt und zerstört werden können. Diese, vor allem in Tierversuchen gewonnene Erkenntnis konnte jedoch in der Vergangenheit nicht zum Nutzen unserer Patienten umgesetzt werden, da immunonkologische Therapieansätze in den letzten Jahrzehnten in der Anwendung beim Menschen stets versagt haben. Daher hat, mit Ausnahme der adjuvanten Interferontherapie, keines dieser Verfahren den Einzug in die klinische Versorgung gefunden. Langzeitüberleben unter guter Lebensqualität war dabei sehr wenigen Patienten vorbehalten. Mit den neuen immunologischen Therapieansätzen wird jedoch sowohl das Langzeitüberleben als auch die Lebensqualität onkologischer Patienten neu definiert. Auf die neuen "Immun-Checkpoint-Inhibitoren" spricht erstmals ein relevanter Teil der behandelten Patienten an und diese zeigen in der Regel langandauernde Remissionen bis hin zur Heilung. Schon jetzt ist klar, dass die Immuntherapie in Zukunft eine der wesentlichen Therapiesäulen bei der Behandlung des metastasierten Melanoms und auch vieler anderer fortgeschrittener Tumoren bilden wird. In dieser Übersicht werden die wichtigsten neuen Therapiemodalitäten besprochen und sowohl deren Wirkprinzip als auch klinische Daten zum Therapieansprechen und zu erwartenden Nebenwirkungen der Therapie referiert.
Collapse
|
5741
|
Blumenthal DT, Yalon M, Vainer GW, Lossos A, Yust S, Tzach L, Cagnano E, Limon D, Bokstein F. Pembrolizumab: first experience with recurrent primary central nervous system (CNS) tumors. J Neurooncol 2016; 129:453-460. [PMID: 27377654 DOI: 10.1007/s11060-016-2190-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 06/23/2016] [Indexed: 12/26/2022]
Abstract
Patients with progressive primary brain tumors (PBT) are attracted to promising new treatments, even prior to convincing data. Anti-PD1 immunotherapies have been in the spotlight since publication of groundbreaking results for metastatic melanoma with pembrolizumab (PBL). Our objective was to report on the response and toxicity of PBL in patients with advanced PBT. We retrospectively reviewed the charts of 22 patients (17 adults and 5 children) with recurrent central nervous system tumors treated with PBL. We analyzed prior antineoplastic therapies, steroid usage, and outcomes. Patients received a median of two neoplastic therapies prior to PBL, and a median of three infusions of PBL in adults and four in children. Twelve patients (9 adults and 3 children) started PBL on steroids (median dose in adults 4 mg; range 2-8, and in children 1.5 mg, range 0.5-4) and five patients received steroids later during PBL treatment. Twelve patients (10 adults and 2 children) received concomitant bevacizumab with PBL. Side effects were minimal. All patients showed progressive tumor growth during therapy. Median OS from the start of PBL was 2.6 months in adults and 3.2 months in children. Two GB patients underwent tumor resection following treatment with PBL. Tumor-lymphocytic response in these cases was unremarkable, and PD-L1 immuno-staining was negative. In this series of 22 patients with recurrent primary brain tumors, PBL showed no clinical or histologic efficacy. We do not recommend further use of PBL for recurrent PBT unless convincing prospective clinical trial data are published.
Collapse
Affiliation(s)
- Deborah T Blumenthal
- Neuro-Oncology Service, Division of Oncology, Tel Aviv Sourasky Medical Center, 64239, Tel Aviv, Israel.
| | - Michal Yalon
- Pediatric Neuro-Oncology Service, Pediatric Hemato-Oncology Department, Chaim Sheba Medical Center, 52621, Tel HaShomer, Israel
| | - Gilad W Vainer
- Molecular Pathology Service, Department of Pathology, Tel Aviv Sourasky Medical Center, 64239, Tel Aviv, Israel
| | - Alexander Lossos
- Leslie and Michael Gaffin Center for Neuro-Oncology, Department of Oncology, Hadassah-Hebrew University Medical Center, 91120, Jerusalem, Israel
| | - Shlomit Yust
- Oncology Institute, Davidoff Center, Rabin Medical Center, 49100, Petah Tikva, Israel
| | - Lior Tzach
- Oncology Institute, Chaim Sheba Medical Center, 52621, Tel HaShomer, Israel
| | - Emanuela Cagnano
- Molecular Pathology Service, Department of Pathology, Tel Aviv Sourasky Medical Center, 64239, Tel Aviv, Israel
| | - Dror Limon
- Oncology Institute, Davidoff Center, Rabin Medical Center, 49100, Petah Tikva, Israel
| | - Felix Bokstein
- Neuro-Oncology Service, Division of Oncology, Tel Aviv Sourasky Medical Center, 64239, Tel Aviv, Israel
| |
Collapse
|
5742
|
Kähler KC, Hassel JC, Heinzerling L, Loquai C, Mössner R, Ugurel S, Zimmer L, Gutzmer R. Nebenwirkungsmanagement bei Immun-Checkpoint-Blockade durch CTLA-4- und PD1-Antikörper beim metastasierten Melanom. J Dtsch Dermatol Ges 2016; 14:662-83. [PMID: 27373240 DOI: 10.1111/ddg.13047_g] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Katharina C Kähler
- Universitätsklinikum Schleswig-Holstein, Campus Kiel, Klinik für Dermatologie, Venerologie und Allergologie
| | - Jessica C Hassel
- Universitätsklinikum Heidelberg, Hautklinik und Nationales Centrum für Tumorerkrankungen
| | | | - Carmen Loquai
- Hautklinik und Poliklinik, Universitätsmedizin Mainz
| | - Rotraut Mössner
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsmedizin Göttingen
| | - Selma Ugurel
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsklinikum Essen
| | - Lisa Zimmer
- Klinik für Dermatologie, Venerologie und Allergologie, Universitätsklinikum Essen
| | - Ralf Gutzmer
- Hauttumorzentrum Hannover, Klinik für Dermatologie, Allergologie und Venerologie, Medizinische Hochschule Hannover
| | | |
Collapse
|
5743
|
Pollack MH, Aston J, Benrashid M, Johnson DB, Puzanov I. Talimogene laherparepvec in advanced melanoma. Expert Opin Orphan Drugs 2016. [DOI: 10.1080/21678707.2016.1186539] [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: 10/21/2022]
Affiliation(s)
- Megan H. Pollack
- Department of Pharmaceutical Services, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, TN, USA
| | - Jonathan Aston
- Department of Pharmaceutical Services, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, TN, USA
| | - Mona Benrashid
- Department of Pharmaceutical Services, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, TN, USA
| | - Douglas B. Johnson
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, TN, USA
| | - Igor Puzanov
- Department of Medicine, Vanderbilt University Medical Center and Vanderbilt Ingram Cancer Center, Nashville, TN, USA
| |
Collapse
|
5744
|
Subbiah V, Maiti A. Headache in a patient with renal cell carcinoma. Eur J Intern Med 2016; 32:e3-4. [PMID: 26905772 DOI: 10.1016/j.ejim.2016.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 02/04/2016] [Indexed: 10/22/2022]
Affiliation(s)
- Vivek Subbiah
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., FC8.3038, Box 0455, Houston, TX 77030, United States.
| | - Abhishek Maiti
- Department of Internal Medicine, The University of Texas Health Science Center at Houston, 6431 Fannin, MSB 1.134, Houston, TX 77030, United States.
| |
Collapse
|
5745
|
Yun S, Vincelette ND, Green MR, Wahner Hendrickson AE, Abraham I. Targeting immune checkpoints in unresectable metastatic cutaneous melanoma: a systematic review and meta-analysis of anti-CTLA-4 and anti-PD-1 agents trials. Cancer Med 2016; 5:1481-91. [PMID: 27167347 PMCID: PMC4867662 DOI: 10.1002/cam4.732] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 03/11/2016] [Accepted: 03/17/2016] [Indexed: 12/18/2022] Open
Abstract
Anti-cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) and anti-programmed cell death-1 (PD-1) inhibitors have been shown to significantly improve survival in patients with metastatic cutaneous melanoma. However, there was some heterogeneity as well as some variation in the degree of benefit across studies. We reviewed randomized trials and performed a meta-analysis to determine the efficacy and safety of immune checkpoint inhibitors in comparison with conventional regimens. Eligible studies were limited to randomized controlled trials comparing anti-CTLA-4 or anti-PD-1 inhibitors to chemotherapy or vaccination treatment in adult patients with unresectable cutaneous metastatic melanoma. Progression-free survival (PFS) rate at 6 months was 28.5% versus 17.7% (RR: 0.84, 95% CI: 0.76-0.93), overall survival (OS) rate at 1 year was 51.2% versus 38.8% (RR: 0.72, 95% CI: 0.59-0.88), and overall response rate (ORR) at 6 months was 29.6% versus 17.7% (RR: 0.85, 95% CI: 0.76-0.95) favoring immune check point inhibitors over chemotherapies or vaccination. Immune check point inhibitors were associated with more frequent immune-related adverse events at 13.7% versus 2.4% of treated patients (RR: 6.74, 95% CI: 4.65-9.75). Subgroup analyses demonstrated significant PFS (RR: 0.92 vs. 0.74, P < 0.00001) and ORR (RR: 0.95 vs. 0.76, P = 0.0004) improvement with anti-PD-1 treatment compared to anti-CTLA-4 when each of them was compared to control treatments. Collectively, these results demonstrate that immune checkpoint inhibitors have superior outcomes compared to conventional chemotherapies or vaccination, and support the results of recent randomized trials that showed superior outcomes with anti-PD-1 agents over ipilimumab in unresectable metastatic cutaneous melanoma patients.
Collapse
Affiliation(s)
- Seongseok Yun
- Department of MedicineUniversity of ArizonaTucsonArizona85721
- Hematology and OncologyUniversity of ArizonaTucsonArizona85721
| | - Nicole D. Vincelette
- Molecular Pharmacology and Experimental TherapeuticsMayo ClinicRochesterMinnesota55905
| | - Myke R. Green
- Hematology and OncologyUniversity of ArizonaTucsonArizona85721
| | | | - Ivo Abraham
- Center for Health Outcomes and PharmacoEconomic ResearchUniversity of ArizonaTucsonArizona85721
- Arizona Cancer CenterUniversity of ArizonaTucsonArizona85721
| |
Collapse
|
5746
|
Franconieri F, Martin-Silva N, de Boysson H, Galateau-Salle F, Emile JF, Bienvenu B, Aouba A. Superior efficacy and tolerance of reduced doses of vemurafenib plus anakinra in Erdheim-Chester disease: Towards the paradigm of combined targeting and immune therapies. Acta Oncol 2016; 55:930-2. [PMID: 27031008 DOI: 10.3109/0284186x.2015.1120885] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Fréderic Franconieri
- Department of Internal Medicine and Clinical Immunology, CHU Côte De Nacre, Université Basse Normandie, Caen, France
| | - Nicolas Martin-Silva
- Department of Internal Medicine and Clinical Immunology, CHU Côte De Nacre, Université Basse Normandie, Caen, France
| | - Hubert de Boysson
- Department of Internal Medicine and Clinical Immunology, CHU Côte De Nacre, Université Basse Normandie, Caen, France
| | | | | | - Boris Bienvenu
- Department of Internal Medicine and Clinical Immunology, CHU Côte De Nacre, Université Basse Normandie, Caen, France
| | - Achille Aouba
- Department of Internal Medicine and Clinical Immunology, CHU Côte De Nacre, Université Basse Normandie, Caen, France
| |
Collapse
|
5747
|
Apigenin inhibits the inducible expression of programmed death ligand 1 by human and mouse mammary carcinoma cells. Cancer Lett 2016; 380:424-433. [PMID: 27378243 DOI: 10.1016/j.canlet.2016.06.023] [Citation(s) in RCA: 83] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 06/26/2016] [Accepted: 06/28/2016] [Indexed: 12/11/2022]
Abstract
Programmed death ligand 1 (PD-L1) is expressed by many cancer cell types, as well as by activated T cells and antigen-presenting cells. Constitutive and inducible PD-L1 expression contributes to immune evasion by breast cancer (BC) cells. We show here that the dietary phytochemical apigenin inhibited interferon (IFN)-γ-induced PD-L1 upregulation by triple-negative MDA-MB-468 BC cells, HER2(+) SK-BR-3 BC cells, and 4T1 mouse mammary carcinoma cells, as well as human mammary epithelial cells, but did not affect constitutive PD-L1 expression by triple-negative MDA-MB-231 BC cells. IFN-β-induced expression of PD-L1 by MDA-MB-468 cells was also inhibited by apigenin. In addition, luteolin, the major metabolite of apigenin, inhibited IFN-γ-induced PD-L1 expression by MDA-MB-468 cells. Apigenin-mediated inhibition of IFN-γ-induced PD-L1 expression by MDA-MB-468 and 4T1 cells was associated with reduced phosphorylation of STAT1, which was early and transient at Tyr701 and sustained at Ser727. Apigenin-mediated inhibition of IFN-γ-induced PD-L1 expression by MDA-MB-468 cells also increased proliferation and interleukin-2 synthesis by PD-1-expressing Jurkat T cells that were co-cultured with MDA-MB-468 cells. Apigenin therefore has the potential to increase the vulnerability of BC cells to T cell-mediated anti-tumor immune responses.
Collapse
|
5748
|
Levy A, Chargari C, Marabelle A, Perfettini JL, Magné N, Deutsch E. Can immunostimulatory agents enhance the abscopal effect of radiotherapy? Eur J Cancer 2016; 62:36-45. [DOI: 10.1016/j.ejca.2016.03.067] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 03/14/2016] [Accepted: 03/15/2016] [Indexed: 12/13/2022]
|
5749
|
Yu H, Boyle TA, Zhou C, Rimm DL, Hirsch FR. PD-L1 Expression in Lung Cancer. J Thorac Oncol 2016; 11:964-75. [PMID: 27117833 PMCID: PMC5353357 DOI: 10.1016/j.jtho.2016.04.014] [Citation(s) in RCA: 334] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 04/11/2016] [Accepted: 04/16/2016] [Indexed: 12/26/2022]
Abstract
Immunotherapies targeted against programmed death ligand 1 (PD-L1) and its receptor (PD-1) have improved survival in a subset of patients with advanced lung cancer. PD-L1 protein expression has emerged as a biomarker that predicts which patients are more likely to respond to immunotherapy. The understanding of PD-L1 as a biomarker is complicated by the history of use of different immunohistochemistry platforms with different PD-L1 antibodies, scoring systems, and positivity cut-offs for immunotherapy clinical trials with different anti-PD-L1 and anti-PD-1 drugs. Herein, we summarize the brief history of PD-L1 as a biomarker and describe the challenges remaining to harmonize PD-L1 detection and interpretation for best patient care.
Collapse
Affiliation(s)
- Hui Yu
- Department of Medicine/Medical Oncology, University of Colorado Cancer Center, University of Colorado, Denver, Colorado
| | - Theresa A Boyle
- Department of Medicine/Medical Oncology, University of Colorado Cancer Center, University of Colorado, Denver, Colorado; Department of Pathology, University of Colorado, Denver, Colorado; Department of Pathology, Moffitt Cancer Center, Tampa, Florida; Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, Florida
| | - Caicun Zhou
- Department of Oncology, Shanghai Pulmonary Hospital, Shanghai, People's Republic of China
| | - David L Rimm
- Department of Pathology, Yale University, New Haven, Connecticut
| | - Fred R Hirsch
- Department of Medicine/Medical Oncology, University of Colorado Cancer Center, University of Colorado, Denver, Colorado; Department of Pathology, University of Colorado, Denver, Colorado.
| |
Collapse
|
5750
|
Carlo MI, Voss MH, Motzer RJ. Checkpoint inhibitors and other novel immunotherapies for advanced renal cell carcinoma. Nat Rev Urol 2016; 13:420-31. [PMID: 27324121 PMCID: PMC5532875 DOI: 10.1038/nrurol.2016.103] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The management of advanced renal cell carcinoma (RCC) has dramatically changed over the past decade. Therapies that target the vascular endothelial growth factor (VEGF) and mammalian target of rapamycin (mTOR) pathways have considerably expanded treatment options; however, most patients with advanced RCC still have limited overall survival. Increased understanding of the mechanisms of T cell-antigen recognition and function has led to the development of novel immunotherapies to treat cancer, chief among them inhibitors of checkpoint receptors - molecules whose function is to restrain the host immune response. In 2015, the FDA approved the first checkpoint inhibitor nivolumab for patients with advanced RCC following treatment with antiangiogenic therapy based on improved overall survival compared with the standard of care. Ongoing phase III trials are comparing checkpoint-inhibitor-based combination regimens with antiangiogenesis agents in the first-line setting. The field is evolving rapidly, with many clinical trials already testing several checkpoint inhibitors alone, in combination, or with other targeted therapies. In addition, different novel immune therapies are being investigated including vaccines, T-cell agonists, and chimeric antigen receptor T cells. Determining which patients will benefit from these therapies and which combination approaches will result in better response will be important as this field evolves.
Collapse
Affiliation(s)
- Maria I Carlo
- Memorial Sloan Kettering Cancer Center 1275 York Avenue New York, New York 10065, USA
| | - Martin H Voss
- Memorial Sloan Kettering Cancer Center 1275 York Avenue New York, New York 10065, USA
| | - Robert J Motzer
- Memorial Sloan Kettering Cancer Center 1275 York Avenue New York, New York 10065, USA
| |
Collapse
|