2451
|
Ito K, Schöder H, Teng R, Humm JL, Ni A, Wolchok JD, Weber WA. Prognostic value of baseline metabolic tumor volume measured on 18F-fluorodeoxyglucose positron emission tomography/computed tomography in melanoma patients treated with ipilimumab therapy. Eur J Nucl Med Mol Imaging 2018; 46:930-939. [DOI: 10.1007/s00259-018-4211-0] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/02/2018] [Indexed: 12/13/2022]
|
2452
|
Nicolson NG, Han D. Desmoplastic melanoma. J Surg Oncol 2018; 119:208-215. [PMID: 30481377 DOI: 10.1002/jso.25317] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 11/11/2018] [Indexed: 12/12/2022]
Abstract
Desmoplastic melanoma (DM) is a rare melanoma variant that has unique biology and pathology compared with conventional melanoma (non-DM). Importantly, DM is classified into pure and mixed histologic subtypes, which have been correlated with outcomes. Management of DM broadly mirrors that of non-DM; however, there are unique considerations for DM that influence treatment approaches. This paper will provide a contemporary overview of this disease and will review the literature regarding the management of DM.
Collapse
Affiliation(s)
- Norman G Nicolson
- Department of Surgery, Section of Surgical Oncology, Yale School of Medicine, New Haven, Connecticut
| | - Dale Han
- Division of Surgical Oncology, Department of Surgery, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
2453
|
Eggermont AMM, Crittenden M, Wargo J. Combination Immunotherapy Development in Melanoma. Am Soc Clin Oncol Educ Book 2018; 38:197-207. [PMID: 30231333 DOI: 10.1200/edbk_201131] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Melanoma has been the most important cancer to drive immunotherapy development of solid tumors. Since 2010, immunotherapy has been revolutionized by the concept of breaking tolerance. It represents a major paradigm shift and marks the beginning of a new era. The impact of the first immune checkpoint inhibitors, anti-CTLA-4 and anti-PD-1/anti-PD-L1, is unprecedented. In 7 years, it transformed advanced-stage melanoma into a curable disease in over 50% of patients. Another major step has been the development of the combination of BRAF inhibitors plus MEK inhibitors in the treatment of BRAF-mutant melanomas. For the treatment of advanced disease, approvals were obtained for the immune checkpoint inhibitors ipilimumab (2011), nivolumab (2014), pembrolizumab (2014), the combination ipilimumab plus nivolumab (2015), and the oncolytic virus vaccine laherparepvec (2015). The combination dabrafenib plus trametinib for BRAF-mutant melanoma was approved in 2014, with similar success for other BRAF plus MEK inhibitor combinations. Because of its unique therapeutic index (high efficacy and low toxicity) anti-PD-1 agents (nivolumab and pembrolizumab) have now been placed at the center of practically all combination therapy development strategies in melanoma. Anti-PD-1 agents are the central molecule for combinations with a great variety of other immunotherapeutics such as immune checkpoint inhibitors, agonists, IDO inhibitors, macrophage polarizing agents, monoclonal antibodies, vaccines, targeted agents, chemotherapeutics, radiation therapy, and even microbiome modulators.
Collapse
Affiliation(s)
- Alexander M M Eggermont
- From the Gustave Roussy Cancer Institute and University Paris-Saclay, Villejuif, France; Earle A. Chiles Research Institute, Portland, OR; The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Marka Crittenden
- From the Gustave Roussy Cancer Institute and University Paris-Saclay, Villejuif, France; Earle A. Chiles Research Institute, Portland, OR; The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jennifer Wargo
- From the Gustave Roussy Cancer Institute and University Paris-Saclay, Villejuif, France; Earle A. Chiles Research Institute, Portland, OR; The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
2454
|
Verma V, Sprave T, Haque W, Simone CB, Chang JY, Welsh JW, Thomas CR. A systematic review of the cost and cost-effectiveness studies of immune checkpoint inhibitors. J Immunother Cancer 2018; 6:128. [PMID: 30470252 PMCID: PMC6251215 DOI: 10.1186/s40425-018-0442-7] [Citation(s) in RCA: 240] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 11/05/2018] [Indexed: 12/26/2022] Open
Abstract
Background Escalating healthcare costs are necessitating the practice of value-based oncology. It is crucial to critically evaluate the economic impact of influential but expensive therapies such as immune checkpoint inhibitors (ICIs). To date, no systematic assessment of the cost-effectiveness (CE) of ICIs has been performed. Methods PRISMA-guided systematic searches of the PubMed database were conducted. Studies of head/neck (n = 3), lung (n = 5), genitourinary (n = 4), and melanoma (n = 8) malignancies treated with ICIs were evaluated. The reference willingness-to-pay (WTP) threshold was $100,000/QALY. Results Nivolumab was not cost-effective over chemotherapy for recurrent/metastatic head/neck cancers (HNCs). For non-small cell lung cancer (NSCLC), nivolumab was not cost-effective for a general cohort, but increased PD-L1 cutoffs resulted in CE. Pembrolizumab was cost-effective for both previously treated and newly-diagnosed metastatic NSCLC. For genitourinary cancers (GUCs, renal cell and bladder cancers), nivolumab and pembrolizumab were not cost-effective options. Regarding metastatic/unresected melanoma, ipilimumab monotherapy is less cost-effective than nivolumab, nivolumab/ipilimumab, and pembrolizumab. The addition of ipilimumab to nivolumab monotherapy was not adequately cost-effective. Pembrolizumab or nivolumab monotherapy offered comparable CE profiles. Conclusions With limited data and from the reference WTP, nivolumab was not cost-effective for HNCs. Pembrolizumab was cost-effective for NSCLC; although not the case for nivolumab, applying PD-L1 cutoffs resulted in adequate CE. Most data for nivolumab and pembrolizumab in GUCs did not point towards adequate CE. Contrary to ipilimumab, either nivolumab or pembrolizumab is cost-effective for melanoma. Despite these conclusions, it cannot be overstated that careful patient selection is critical for CE. Future publication of CE investigations and clinical trials (along with longer follow-up of existing data) could substantially alter conclusions from this analysis.
Collapse
Affiliation(s)
- Vivek Verma
- Department of Radiation Oncology, Allegheny General Hospital, 320 East North Ave, Pittsburgh, PA, 15212, USA.
| | - Tanja Sprave
- Department of Radiation Oncology, University Hospital Heidelberg, Heidelberg, Germany
| | - Waqar Haque
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, TX, USA
| | - Charles B Simone
- Department of Radiation Oncology, University of Maryland Medical Center, Baltimore, MD, USA
| | - Joe Y Chang
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - James W Welsh
- Department of Radiation Oncology, University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - Charles R Thomas
- Department of Radiation Oncology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
2455
|
Micevic G, Thakral D, McGeary M, Bosenberg MW. PD-L1 methylation regulates PD-L1 expression and is associated with melanoma survival. Pigment Cell Melanoma Res 2018; 32:435-440. [PMID: 30343532 DOI: 10.1111/pcmr.12745] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 08/30/2018] [Accepted: 10/01/2018] [Indexed: 11/27/2022]
Abstract
The aim of this study is to determine the significance of programmed death ligand 1 (PD-L1 or CD274) methylation in relation to PD-L1 expression and survival in melanoma. Despite the clinical importance of therapies targeting the PD-1/PD-L1 immune checkpoint in melanoma, factors regulating PD-L1 expression, including epigenetic mechanisms, are not completely understood. In this study, we examined PD-L1 promoter methylation in relation to PD-L1 expression and overall survival in melanoma patients. Our results suggest that DNA methylation regulates PD-L1 expression in melanoma, and we identify the key methylated CpG loci in the PD-L1 promoter, establish PD-L1 methylation as an independent survival prognostic factor, provide proof of concept for altering PD-L1 expression by hypomethylating agents, and uncover that PD-L1 methylation is associated with an interferon signaling transcriptional phenotype. Based on our findings, measuring and altering PD-L1 promoter DNA methylation may have potential prognostic and therapeutic applications in melanoma.
Collapse
Affiliation(s)
- Goran Micevic
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut.,Department of Dermatology, Yale School of Medicine, New Haven, Connecticut
| | - Durga Thakral
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut
| | - Meaghan McGeary
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Marcus W Bosenberg
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut.,Department of Dermatology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
2456
|
Fujimura T, Sato Y, Tanita K, Kambayashi Y, Otsuka A, Fujisawa Y, Yoshino K, Matsushita S, Funakoshi T, Hata H, Yamamoto Y, Uchi H, Nonomura Y, Tanaka R, Aoki M, Imafuku K, Okuhira H, Wada N, Irie H, Hidaka T, Hashimoto A, Aiba S. Serum Level of Soluble CD163 May Be a Predictive Marker of the Effectiveness of Nivolumab in Patients With Advanced Cutaneous Melanoma. Front Oncol 2018; 8:530. [PMID: 30510916 PMCID: PMC6252386 DOI: 10.3389/fonc.2018.00530] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 10/30/2018] [Indexed: 12/17/2022] Open
Abstract
Antibodies against programmed cell death protein 1, such as nivolumab and pembrolizumab, are widely used for treating various cancers, including advanced melanoma. Nivolumab significantly prolongs survival in patients with metastatic melanoma, and sequential administration with lipilimumab may improve outcomes when switched at the appropriate time. Biomarkers are therefore needed to evaluate nivolumab efficacy soon after first administration. This study analyzed serum levels of soluble cluster of differentiation 163 (sCD163) in 59 cases of advanced cutaneous melanoma and 16 cases of advanced mucosal melanoma treated using nivolumab. Serum levels of sCD163 were significantly increased after 6 weeks in responders compared to non-responders after initial administration of nivolumab for cutaneous melanoma. In contrast, no significant difference between responders and non-responders was seen among patients with non-cutaneous melanoma. These results suggest that sCD163 may be useful as a biomarker for selecting patients with advanced cutaneous melanoma most likely to benefit from anti-melanoma immunotherapy.
Collapse
Affiliation(s)
- Taku Fujimura
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yota Sato
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kayo Tanita
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yumi Kambayashi
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Atsushi Otsuka
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yasuhiro Fujisawa
- Department of Dermatology, Faculty of University of Tsukuba, Tsukuba, Japan
| | - Koji Yoshino
- Department of Dermatology, Tokyo Metropolitan Cancer and Infectious Disease Center Komagome Hospital, Tokyo, Japan
| | - Shigeto Matsushita
- Department of Dermato-Oncology, Dermatology, National Hospital Organization Kagoshima Medical Center, Kagoshima, Japan
| | - Takeru Funakoshi
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Hiroo Hata
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yuki Yamamoto
- Department of Dermatology, Wakayama Medical University, Wakayama, Japan
| | - Hiroshi Uchi
- Department of Dermatology, Kyushu University Graduate School of Medicine, Fukuoka, Japan
| | - Yumi Nonomura
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryota Tanaka
- Department of Dermatology, Faculty of University of Tsukuba, Tsukuba, Japan
| | - Megumi Aoki
- Department of Dermato-Oncology, Dermatology, National Hospital Organization Kagoshima Medical Center, Kagoshima, Japan
| | - Keisuke Imafuku
- Department of Dermatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hisako Okuhira
- Department of Dermatology, Wakayama Medical University, Wakayama, Japan
| | - Naoko Wada
- Department of Dermatology, Kyushu University Graduate School of Medicine, Fukuoka, Japan
| | - Hiroyuki Irie
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takanori Hidaka
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akira Hashimoto
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Setsuya Aiba
- Department of Dermatology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
2457
|
Shi G, Yang Q, Zhang Y, Jiang Q, Lin Y, Yang S, Wang H, Cheng L, Zhang X, Li Y, Wang Q, Liu Y, Wang Q, Zhang H, Su X, Dai L, Liu L, Zhang S, Li J, Li Z, Yang Y, Yu D, Wei Y, Deng H. Modulating the Tumor Microenvironment via Oncolytic Viruses and CSF-1R Inhibition Synergistically Enhances Anti-PD-1 Immunotherapy. Mol Ther 2018; 27:244-260. [PMID: 30527756 DOI: 10.1016/j.ymthe.2018.11.010] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 11/05/2018] [Accepted: 11/08/2018] [Indexed: 02/05/2023] Open
Abstract
Immunotherapy based on the immune checkpoint blockade has emerged as the most promising approach for cancer therapy. However, the proportion of colorectal cancer patients who benefit from immunotherapy is small due to the immunosuppressive tumor microenvironment. Hence, combination immunotherapy is an ideal strategy to overcome this limitation. In this study, we developed a novel combination of CSF-1R (colony-stimulating factor 1 receptor) inhibitor (PLX3397), oncolytic viruses, and anti-PD-1 antibody. Our results demonstrated that the triple treatment synergistically conferred significant tumor control and prolonged the survival of mouse models of colon cancer. Approximately 43% and 82% of mice bearing the CT26 and MC38 tumor, respectively, survived long term following the triple treatment. This combination therapy reprogrammed the immunosuppressive tumor microenvironment toward a CD8+ T cell-biased anti-tumor immunity by increasing T cell infiltration in the tumor and augmenting anti-tumor CD8+ T cell function. Our results provide a robust strategy for clinical combination therapy.
Collapse
Affiliation(s)
- Gang Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Qianmei Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Yujing Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Qingyuan Jiang
- Department of Obstetrics, Sichuan Provincial Hospital for Women and Children, Chengdu, China
| | - Yi Lin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Shenshen Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Huiling Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Lin Cheng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Xin Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Yimin Li
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Qingnan Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Yi Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Qin Wang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Hantao Zhang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Xiaolan Su
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Lei Liu
- Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shuang Zhang
- Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jia Li
- Innovent Biologics, Inc., Suzhou, Jiangsu, China
| | - Zhi Li
- Innovent Biologics, Inc., Suzhou, Jiangsu, China
| | - Yang Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Dechao Yu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China; Innovent Biologics, Inc., Suzhou, Jiangsu, China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China.
| |
Collapse
|
2458
|
Nie RC, Chen FP, Yuan SQ, Luo YS, Chen S, Chen YM, Chen XJ, Chen YB, Li YF, Zhou ZW. Evaluation of objective response, disease control and progression-free survival as surrogate end-points for overall survival in anti-programmed death-1 and anti-programmed death ligand 1 trials. Eur J Cancer 2018; 106:1-11. [PMID: 30453169 DOI: 10.1016/j.ejca.2018.10.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND We aimed to assess whether the Response Evaluation Criteria in Solid Tumors (RECIST) criteria-based objective response rate (ORR), disease control rate (DCR) and progression-free survival (PFS) could be valid surrogate end-points for overall survival (OS) in anti-programmed death-1 (PD-1)/programmed death ligand 1 (PD-L1) trials. METHODS We systematically reviewed phase 2 and phase 3 trials of anti-PD-1/PD-L1 drug trials of advanced or recurrent solid tumours that reported OS and at least one of the RECIST criteria-based end-points. We used Spearman rank correlation to evaluate the strength of the association between these end-points and OS and a linear regression model, weighted by the sample size, to assess the association between the treatment effect on these end-points and OS. We also performed sensitivity analyses and a leave-one-out cross-validation approach to evaluate the robustness of our findings. RESULTS Forty-three qualifying trails comprising 15,088 patients were eligible. PFS showed good correlation with OS (squared Spearman rank correlation coefficient [rs2] = 0.54; P < 0.001), while ORR and DCR illustrated moderate association with OS (rs2 = 0.29 and 0.28, respectively; both P < 0.001). The correlation was moderate between the treatment effects on PFS and OS (coefficient of determination [R2] = 0.37, P < 0.001) and poor among ORR, DCR and OS (R2 = 0.10 and 0.08, respectively); these were confirmed by sensitivity analyses (all R2 < 0.75) and the leave-one-out cross-validation approach. CONCLUSIONS No RECIST criteria-based end-points could be a valid surrogate for OS. At present, we proposed to set OS as the primary end-point in anti-PD-1/PD-L1 drug trials of advanced or recurrent solid tumours.
Collapse
Affiliation(s)
- Run-Cong Nie
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Fo-Ping Chen
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Shu-Qiang Yuan
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ying-Shan Luo
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Shi Chen
- Department of Gastric Surgery, The 6th Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yong-Ming Chen
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xiao-Jiang Chen
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Ying-Bo Chen
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yuan-Fang Li
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Zhi-Wei Zhou
- Department of Gastric Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| |
Collapse
|
2459
|
Rådestad E, Klynning C, Stikvoort A, Mogensen O, Nava S, Magalhaes I, Uhlin M. Immune profiling and identification of prognostic immune-related risk factors in human ovarian cancer. Oncoimmunology 2018; 8:e1535730. [PMID: 30713791 PMCID: PMC6343785 DOI: 10.1080/2162402x.2018.1535730] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 09/25/2018] [Accepted: 10/04/2018] [Indexed: 12/15/2022] Open
Abstract
Suppression of immune reactivity by increased expression of co-inhibitory receptors has been discussed as a major reason as to why the immune system fails to control tumor development. Elucidating the co-inhibitory expression pattern of tumor-infiltrating lymphocytes in different cancer types will help to develop future treatment strategies. We characterized markers reflecting and affecting T-cell functionality by flow cytometry on lymphocytes isolated from blood, ascites and tumor from advanced ovarian cancer patients (n = 35). Significantly higher proportions of CD4+ and CD8+ T-cells expressed co-inhibitory receptors LAG-3, PD-1 and TIM-3 in tumor and ascites compared to blood. Co-expression was predominantly observed among intratumoral CD8+ T-cells and the most common combination was PD-1 and TIM-3. Analysis of 26 soluble factors revealed highest concentrations of IP-10 and MCP-1 in both ascites and tumor. Correlating these results with clinical outcome revealed the proportion of CD8+ T-cells without expression of LAG-3, PD-1 and TIM-3 to be beneficial for overall survival. In total we identified eight immune-related risk factors associated with reduced survival. Ex vivo activation showed tumor-derived CD4+ and CD8+ T-cells to be functionally active, assessed by the production of IFN-γ, IL-2, TNF-α, IL-17 and CD107a. Blocking the PD-1 receptor resulted in significantly increased release of IFN-γ suggesting potential reinvigoration. The ovarian tumor environment exhibits an inflammatory milieu with abundant presence of infiltrating immune cells expressing inhibitory checkpoints. Importantly, we found subsets of CD8+ T-cells with double and triple expression of co-inhibitory receptors, supporting the need for multiple checkpoint-targeting agents to overcome T-cell dysfunction in ovarian cancer.
Collapse
Affiliation(s)
- Emelie Rådestad
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Charlotte Klynning
- Department of Gynecological Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Arwen Stikvoort
- Department of Hematology, VU University Medical Center/Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Ole Mogensen
- Division of Obstetrics and Gynecology, Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Silvia Nava
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Isabelle Magalhaes
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Michael Uhlin
- Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden.,Department of Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden.,Department of Applied Physics, Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
2460
|
Barroso-Sousa R, Ott PA. Transformation of Old Concepts for a New Era of Cancer Immunotherapy: Cytokine Therapy and Cancer Vaccines as Combination Partners of PD1/PD-L1 Inhibitors. Curr Oncol Rep 2018; 21:1. [PMID: 30498900 DOI: 10.1007/s11912-018-0738-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW Immune checkpoint inhibitors (ICI) are only effective in a subset of patients. Here, we will review the rationale and data supporting the combination of PD-1 pathway inhibition with recombinant cytokines and neoantigen-based cancer vaccines that can potentially increase the number of patients who will benefit from immunotherapy. RECENT FINDINGS The safety and tolerability of new interleukin(IL)-2 formulations, IL-15 super agonist, and PEGylated IL-10 have been evaluated in early phase clinical trials with promising efficacy data, both as monotherapy and in combination with ICI. Larger studies focusing on the efficacy of these combinations are ongoing. Personalized neoantigen-based cancer vaccines, enabled by improvements in sequencing computational capabilities, have been proven to be feasible, safe, and able to trigger a consistent vaccine-specific immune response in cancer patients. New pharmacologically modified recombinant cytokines and personalized neoantigen-based vaccines may turn these approaches into powerful tools for effective combination immunotherapy.
Collapse
Affiliation(s)
- Romualdo Barroso-Sousa
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Dana 2127, Boston, MA, 02215, USA
| | - Patrick A Ott
- Department of Medical Oncology, Dana-Farber Cancer Institute, 450 Brookline Avenue, Dana 2127, Boston, MA, 02215, USA.
| |
Collapse
|
2461
|
Luther C, Swami U, Zhang J, Milhem M, Zakharia Y. Advanced stage melanoma therapies: Detailing the present and exploring the future. Crit Rev Oncol Hematol 2018; 133:99-111. [PMID: 30661664 DOI: 10.1016/j.critrevonc.2018.11.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 08/07/2018] [Accepted: 11/07/2018] [Indexed: 12/21/2022] Open
Abstract
Metastatic melanoma therapies have proliferated over the last ten years. Prior to this, decades passed with only very few drugs available to offer our patients, and even then, those few drugs had minimal survival benefits. Many treatment options emerged over the last ten years with diverse mechanisms of action. Further, combination regimens have demonstrated superiority over monotherapy, especially for targeted agents. Each therapeutic combination possesses different advantages and side effect profiles. In this review, we outline the United States Food and Drug Administration-approved melanoma treatment agents and therapies currently in clinical development, focusing on combination approaches.
Collapse
Affiliation(s)
- Chelsea Luther
- Department of Dermatology, Henry Ford Hospital, Detroit, MI, United States
| | - Umang Swami
- Department of Internal Medicine, Division of Hematology, Oncology and Blood and Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Jun Zhang
- Department of Internal Medicine, Division of Hematology, Oncology and Blood and Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Mohammed Milhem
- Department of Internal Medicine, Division of Hematology, Oncology and Blood and Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, United States
| | - Yousef Zakharia
- Department of Internal Medicine, Division of Hematology, Oncology and Blood and Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, United States.
| |
Collapse
|
2462
|
Shifrut E, Carnevale J, Tobin V, Roth TL, Woo JM, Bui CT, Li PJ, Diolaiti ME, Ashworth A, Marson A. Genome-wide CRISPR Screens in Primary Human T Cells Reveal Key Regulators of Immune Function. Cell 2018; 175:1958-1971.e15. [PMID: 30449619 DOI: 10.1016/j.cell.2018.10.024] [Citation(s) in RCA: 367] [Impact Index Per Article: 52.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/08/2018] [Accepted: 10/08/2018] [Indexed: 12/11/2022]
Abstract
Human T cells are central effectors of immunity and cancer immunotherapy. CRISPR-based functional studies in T cells could prioritize novel targets for drug development and improve the design of genetically reprogrammed cell-based therapies. However, large-scale CRISPR screens have been challenging in primary human cells. We developed a new method, single guide RNA (sgRNA) lentiviral infection with Cas9 protein electroporation (SLICE), to identify regulators of stimulation responses in primary human T cells. Genome-wide loss-of-function screens identified essential T cell receptor signaling components and genes that negatively tune proliferation following stimulation. Targeted ablation of individual candidate genes characterized hits and identified perturbations that enhanced cancer cell killing. SLICE coupled with single-cell RNA sequencing (RNA-seq) revealed signature stimulation-response gene programs altered by key genetic perturbations. SLICE genome-wide screening was also adaptable to identify mediators of immunosuppression, revealing genes controlling responses to adenosine signaling. The SLICE platform enables unbiased discovery and characterization of functional gene targets in primary cells.
Collapse
Affiliation(s)
- Eric Shifrut
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Julia Carnevale
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Victoria Tobin
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Theodore L Roth
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jonathan M Woo
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Christina T Bui
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - P Jonathan Li
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Morgan E Diolaiti
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Alan Ashworth
- UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alexander Marson
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA; UCSF Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
2463
|
Tarhini AA, Frankel P, Ruel C, Ernstoff MS, Kuzel TM, Logan TF, Khushalani NI, Tawbi HA, Margolin KA, Awasthi S, Butterfield LH, McDermott D, Chen A, Lara PN, Kirkwood JM. NCI 8628: A randomized phase 2 study of ziv-aflibercept and high-dose interleukin 2 or high-dose interleukin 2 alone for inoperable stage III or IV melanoma. Cancer 2018; 124:4332-4341. [PMID: 30303516 PMCID: PMC6504933 DOI: 10.1002/cncr.31734] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 08/22/2016] [Accepted: 10/31/2016] [Indexed: 11/09/2022]
Abstract
BACKGROUND Interleukin 2 (IL-2) is a growth factor for T and natural killer cells, promotes proinflammatory cytokines, and can lead to durable responses in patients with melanoma. Vascular endothelial growth factor (VEGF) promotes angiogenesis and modulates host innate and adaptive immunity. High VEGF levels were found to be associated with nonresponse to IL-2. Ziv-aflibercept may deplete VEGF and thereby enhance antitumor T-cell responses, thus supporting a combination immunotherapeutic strategy with IL-2. METHODS NCI 8628 was a phase 2 trial of ziv-aflibercept and IL-2 (arm A) versus IL-2 alone (arm B) randomized at 2:1, respectively. Eligible patients had inoperable American Joint Committee on Cancer stage III or stage IV melanoma. The primary endpoint was progression-free survival (PFS). RESULTS A total of 89 patients were enrolled and 84 patients were treated. The median follow-up was 41.4 months. Among treated patients (55 patients in arm A and 29 patients in arm B), PFS was significantly improved in favor of arm A, with a median of 6.9 months (95% confidence interval [95% CI], 4.1-8.7 months) versus 2.3 months (95% CI, 1.6-3.5 months) (P<.001). No significant difference was noted with regard to overall survival, with a median of 26.9 months (95% CI, 14.4-63.6 months) for arm A and 24.2 months (95% CI, 11.3-36.4 months) for arm B. The response rate (according to Response Evaluation Criteria In Solid Tumors [RECIST]) was 22% in arm A (4 complete responses [CRs] and 8 partial responses [PRs]) and 17% in arm B (1 CR and 4 PRs). Stable disease or PR or CR was noted in 65% of patients in arm A and 48% of patients in arm B. The combination was found to be superior to monotherapy in patients with high and low levels of serum VEGF and VEGF receptor 2. Adverse events were consistent with the expected profiles of monotherapy with IL-2 and ziv-aflibercept. CONCLUSIONS Ziv-aflibercept and IL-2 were found to significantly improve PFS compared with IL-2 alone, thereby meeting the primary endpoint of the current study. These findings support further study of immunotherapeutic combination strategies involving VEGF inhibitors.
Collapse
Affiliation(s)
- Ahmad A. Tarhini
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Cleveland Clinic and Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Paul Frankel
- City of Hope National Medical Center, Duarte, CA, USA
| | | | | | | | | | | | - Hussein A. Tawbi
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Lisa H. Butterfield
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA
- Departments of Medicine, Surgery and Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Alice Chen
- Cancer Therapy Evaluation Program (CTEP), National Cancer Institute, Bethesda, MD, USA
| | - Primo N. Lara
- University of California Davis Comprehensive Cancer Center, Sacramento, CA, USA
| | | |
Collapse
|
2464
|
Duperret EK, Trautz A, Stoltz R, Patel A, Wise MC, Perales-Puchalt A, Smith T, Broderick KE, Masteller E, Kim J, Humeau L, Muthumani K, Weiner DB. Synthetic DNA-Encoded Monoclonal Antibody Delivery of Anti-CTLA-4 Antibodies Induces Tumor Shrinkage In Vivo. Cancer Res 2018; 78:6363-6370. [PMID: 30287678 PMCID: PMC6239932 DOI: 10.1158/0008-5472.can-18-1429] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 08/16/2018] [Accepted: 09/17/2018] [Indexed: 01/07/2023]
Abstract
Antibody-based immune therapies targeting the T-cell checkpoint molecules CTLA-4 and PD-1 have affected cancer therapy. However, this immune therapy requires complex manufacturing and frequent dosing, limiting the global use of this treatment. Here, we focused on the development of a DNA-encoded monoclonal antibody (DMAb) approach for delivery of anti-CTLA-4 monoclonal antibodies in vivo With this technology, engineered and formulated DMAb plasmids encoding IgG inserts were directly injected into muscle and delivered intracellularly by electroporation, leading to in vivo expression and secretion of the encoded IgG. DMAb expression from a single dose can continue for several months without the need for repeated administration. Delivery of an optimized DMAb encoding anti-mouse CTLA-4 IgG resulted in high serum levels of the antibody as well as tumor regression in Sa1N and CT26 tumor models. DNA-delivery of the anti-human CTLA-4 antibodies ipilimumab and tremelimumab in mice achieved potent peak levels of approximately 85 and 58 μg/mL, respectively. These DMAb exhibited prolonged expression, with maintenance of serum levels at or above 15 μg/mL for over a year. Anti-human CTLA-4 DMAbs produced in vivo bound to human CTLA-4 protein expressed on stimulated human peripheral blood mononuclear cells and induced T-cell activation in a functional assay ex vivo In summary, direct in vivo expression of DMAb encoding checkpoint inhibitors serves as a novel tool for immunotherapy that could significantly improve availability and provide broader access to such therapies.Significance: DNA-encoded monoclonal antibodies represent a novel technology for delivery and expression of immune checkpoint blockade antibodies, thus expanding patient access to, and possible clinical applications of, these therapies. Cancer Res; 78(22); 6363-70. ©2018 AACR.
Collapse
Affiliation(s)
| | - Aspen Trautz
- The Wistar Institute, Vaccine & Immunotherapy Center, Philadelphia PA
| | - Regina Stoltz
- The Wistar Institute, Vaccine & Immunotherapy Center, Philadelphia PA
| | - Ami Patel
- The Wistar Institute, Vaccine & Immunotherapy Center, Philadelphia PA
| | | | | | | | | | | | | | | | - Kar Muthumani
- The Wistar Institute, Vaccine & Immunotherapy Center, Philadelphia PA
| | - David B. Weiner
- The Wistar Institute, Vaccine & Immunotherapy Center, Philadelphia PA,Corresponding author: David B. Weiner, Vaccine & Immunotherapy Center, The Wistar Institute, 3601 Spruce Street, Philadelphia, PA 19104,
| |
Collapse
|
2465
|
Namikawa K, Kiyohara Y, Takenouchi T, Uhara H, Uchi H, Yoshikawa S, Takatsuka S, Koga H, Wada N, Minami H, Hatsumichi M, Asada S, Namba Y, Yamazaki N. Efficacy and safety of nivolumab in combination with ipilimumab in Japanese patients with advanced melanoma: An open-label, single-arm, multicentre phase II study. Eur J Cancer 2018; 105:114-126. [PMID: 30447539 DOI: 10.1016/j.ejca.2018.09.025] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 09/23/2018] [Indexed: 12/19/2022]
Abstract
AIM The aim of the study was to evaluate the efficacy and safety of nivolumab combined with ipilimumab in treatment-naïve Japanese patients with advanced melanoma. METHODS In this multicentre, single-arm study, treatment-naïve Japanese patients with unresectable stage III/IV or recurrent melanoma received nivolumab (1 mg/kg) plus ipilimumab (3 mg/kg) every 3 weeks for four doses, followed by biweekly doses of nivolumab (3 mg/kg). The primary end-point was centrally assessed objective response rate (ORR). Secondary end-points included overall survival (OS), progression-free survival (PFS), disease control rate and safety. RESULTS The subtypes of the thirty patients enrolled were: 12, mucosal; eight, non-acral cutaneous; seven, acral; two, uveal and one, unknown primary melanoma. The ORR was 43.3% (95% confidence interval [CI]: 25.5, 62.6) with central and local assessment. The centrally and locally assessed disease control rate (95% CI) were 73.3% (54.1, 87.7) and 86.7% (69.3, 96.2), respectively. At the median follow-up period of 14.1 months (range 5.2-27.7), median OS and centrally assessed PFS were not reached. OS (95% CI) at 6, 12, 18 and 24 months was 93.3% (75.9, 98.3), 83.3% (64.5, 92.7), 72.9% (50.0, 86.5) and 65.6% (40.4, 82.2), respectively. Treatment-related adverse events (AEs) occurred in all patients. Grade III-IV and serious AEs occurred, mostly during the combination phase, in 23 (76.7%) and 20 (66.7%) patients, respectively. No treatment-related deaths occurred. CONCLUSIONS This study confirmed the efficacy and safety of nivolumab plus ipilimumab in treatment-naïve Japanese patients with advanced melanoma including rare subtypes. Incidence rates for grade III-IV AEs were high but manageable with appropriate medical attention and treatment. TRIAL REGISTRATION JapicCTI-152869.
Collapse
Affiliation(s)
- Kenjiro Namikawa
- Department of Dermatologic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| | - Yoshio Kiyohara
- Dermatology Division, Shizuoka Cancer Center Hospital, 1007 Shimonagakubo, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan.
| | - Tatsuya Takenouchi
- Department of Dermatology, Niigata Cancer Center Hospital, 2-15-3 Kawagishi-cho, Niigata 961-8566, Japan.
| | - Hisashi Uhara
- Department of Dermatology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan.
| | - Hiroshi Uchi
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | - Shusuke Yoshikawa
- Dermatology Division, Shizuoka Cancer Center Hospital, 1007 Shimonagakubo, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan.
| | - Sumiko Takatsuka
- Department of Dermatology, Niigata Cancer Center Hospital, 2-15-3 Kawagishi-cho, Niigata 961-8566, Japan.
| | - Hiroshi Koga
- Department of Dermatology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan.
| | - Naoko Wada
- Department of Dermatology, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan.
| | - Hironobu Minami
- Department Medical Oncology/Hematology, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan.
| | - Masahiro Hatsumichi
- Ono Pharmaceutical Co. Ltd., 1-8-2 Kyutaromachi, Chuo-ku, Osaka 541-8564, Japan.
| | - Suguru Asada
- Ono Pharmaceutical Co. Ltd., 1-8-2 Kyutaromachi, Chuo-ku, Osaka 541-8564, Japan.
| | - Yoshinobu Namba
- Ono Pharmaceutical Co. Ltd., 1-8-2 Kyutaromachi, Chuo-ku, Osaka 541-8564, Japan.
| | - Naoya Yamazaki
- Department of Dermatologic Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| |
Collapse
|
2466
|
LaRocca CJ, Warner SG. Oncolytic viruses and checkpoint inhibitors: combination therapy in clinical trials. Clin Transl Med 2018; 7:35. [PMID: 30426287 PMCID: PMC6234197 DOI: 10.1186/s40169-018-0214-5] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 10/25/2018] [Indexed: 12/15/2022] Open
Abstract
Advances in the understanding of cancer immunotherapy and the development of multiple checkpoint inhibitors have dramatically changed the current landscape of cancer treatment. Recent large-scale phase III trials (e.g. PHOCUS, OPTiM) are establishing use of oncolytic viruses as another tool in the cancer therapeutics armamentarium. These viruses do not simply lyse cells to achieve their cancer-killing effects, but also cause dramatic changes in the tumor immune microenvironment. This review will highlight the major vector platforms that are currently in development (including adenoviruses, reoviruses, vaccinia viruses, herpesviruses, and coxsackieviruses) and how they are combined with checkpoint inhibitors. These vectors employ a variety of engineered capsid modifications to enhance infectivity, genome deletions or promoter elements to confer selective replication, and encode a variety of transgenes to enhance anti-tumor or immunogenic effects. Pre-clinical and clinical data have shown that oncolytic vectors can induce anti-tumor immunity and markedly increase immune cell infiltration (including cytotoxic CD8+ T cells) into the local tumor microenvironment. This "priming" by the viral infection can change a 'cold' tumor microenvironment into a 'hot' one with the influx of a multitude of immune cells and cytokines. This alteration sets the stage for subsequent checkpoint inhibitor delivery, as they are most effective in an environment with a large lymphocytic infiltrate. There are multiple ongoing clinical trials that are currently combining oncolytic viruses with checkpoint inhibitors (e.g. CAPTIVE, CAPRA, and Masterkey-265), and the initial results are encouraging. It is clear that oncolytic viruses and checkpoint inhibitors will continue to evolve together as a combination therapy for multiple types of cancers.
Collapse
Affiliation(s)
- Christopher J LaRocca
- Division of Surgical Oncology, Department of Surgery, City of Hope National Medical Center, 1500 E Duarte Road, Duarte, CA, 91010, USA
| | - Susanne G Warner
- Division of Surgical Oncology, Department of Surgery, City of Hope National Medical Center, 1500 E Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
2467
|
Jansen CS, Prokhnevska N, Kissick HT. The requirement for immune infiltration and organization in the tumor microenvironment for successful immunotherapy in prostate cancer. Urol Oncol 2018; 37:543-555. [PMID: 30446449 DOI: 10.1016/j.urolonc.2018.10.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 10/01/2018] [Accepted: 10/03/2018] [Indexed: 12/14/2022]
Abstract
Immunotherapy-particularly immune checkpoint blockade-has seen great success in many tumor types. However, checkpoint-based therapies have not demonstrated high levels of success in prostate cancer, and there is much to be learned from both the successes and failures of these treatments. Here we review the evidence that composition of infiltrating immune cells in the tumor microenvironment is fundamental to the response to immunotherapy. Additionally, we discuss the emerging idea that the organization of these immune cells may also be crucial to this response. In prostate cancer, the composition and organization of the tumor immune microenvironment are preeminent topics of discussion and areas of important future investigation.
Collapse
Affiliation(s)
| | | | - Haydn T Kissick
- Department of Urology, Emory University, Atlanta, GA; Department of Microbiology and Immunology, Emory University, Atlanta, GA.
| |
Collapse
|
2468
|
Yentz S, Smith D. Indoleamine 2,3-Dioxygenase (IDO) Inhibition as a Strategy to Augment Cancer Immunotherapy. BioDrugs 2018; 32:311-317. [PMID: 29980987 DOI: 10.1007/s40259-018-0291-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Indoleamine 2,3-dioxygenase (IDO) is an enzyme of interest in immuno-oncology because of the immunosuppressive effects that result from its role in tryptophan catabolism. IDO is upregulated in malignancy and is associated with poor prognosis in multiple cancer types. IDO inhibitors have been developed to target IDO, both directly and indirectly. Pre-clinical data have shown combined IDO and checkpoint inhibition to be an efficacious strategy for tumor control. Clinical trials of IDO inhibitors with chemotherapy or immunotherapy are currently underway. This review describes the function of IDO and its inhibitors and summarizes the efficacy and toxicity data from recent clinical trials with these drugs.
Collapse
Affiliation(s)
- Sarah Yentz
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan Health System, 1500 E. Medical Center Drive, C369 Med Inn Building, SPC 5848, Ann Arbor, MI, 48109, USA.
| | - David Smith
- Division of Hematology/Oncology, Department of Internal Medicine, University of Michigan Health System, 1500 E. Medical Center Drive, C369 Med Inn Building, SPC 5848, Ann Arbor, MI, 48109, USA
| |
Collapse
|
2469
|
Hong B, Cai L, Wang J, Liu S, Zhou J, Ma K, Zhang J, Zhou B, Peng X, Zhang N, Gong K. Differential Expression of PD-L1 Between Sporadic and VHL-Associated Hereditary Clear-Cell Renal Cell Carcinoma and Its Correlation With Clinicopathological Features. Clin Genitourin Cancer 2018; 17:97-104.e1. [PMID: 30522901 DOI: 10.1016/j.clgc.2018.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 10/30/2018] [Accepted: 11/04/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Programmed death ligand-1 (PD-L1) is a potential predictive biomarker for immunotherapy in several malignancies. However, the expression level and clinical significance of PD-L1 in von Hippel-Lindau (VHL)-associated hereditary clear-cell renal cell carcinoma (ccRCC) remain unclear. PATIENTS AND METHODS Surgical specimens were recruited from 129 patients with sporadic ccRCC and 26 patients with VHL-associated hereditary ccRCC. The PD-L1 expression level was assessed using immunohistochemistry. Correlations between PD-L1 expression and clinicopathological features were analyzed. RESULTS In sporadic ccRCC, the positive expression rate of PD-L1 was 47.3% (61/129). Positive PD-L1 expression was correlated with advanced tumor T stage (P = .011), higher Fuhrman nuclear grade (P = .022), poor disease-free survival (P = .037), and sex (P = .025). In the VHL-associated hereditary ccRCC, positive PD-L1 expression rate was 34.6% (9/26), lower than that in sporadic ccRCC. Positive PD-L1 was correlated with higher Fuhrman nuclear grade (P = .008), but not with sex, age, tumor stage, or the onset age of VHL-associated tumors. CONCLUSION Positive PD-L1 expression was correlated with the aggressive clinicopathological features in sporadic and VHL-associated hereditary ccRCC. Whether PD-L1 expression level in ccRCC is related to the effectiveness of programmed death-1/PD-L1 checkpoint inhibitor immunotherapy needs to be further investigated.
Collapse
Affiliation(s)
- Baoan Hong
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China
| | - Lin Cai
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China
| | - Jiangyi Wang
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China
| | - Shengjie Liu
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China
| | - Jingcheng Zhou
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China
| | - Kaifang Ma
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China
| | - Jiufeng Zhang
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China
| | - Bowen Zhou
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China
| | - Xiang Peng
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China
| | - Ning Zhang
- Department of Urology, Beijing Cancer Hospital, Beijing, China.
| | - Kan Gong
- Department of Urology, Peking University First Hospital, Beijing, China; Institute of Urology, Peking University, Beijing, China; National Urological Cancer Center, Beijing, China.
| |
Collapse
|
2470
|
Kokolus KM, Haley JS, Koubek EJ, Gowda R, Dinavahi SS, Sharma A, Claxton DF, Helm KF, Drabick JJ, Robertson GP, Neighbors JD, Hohl RJ, Schell TD. Schweinfurthin natural products induce regression of murine melanoma and pair with anti-PD-1 therapy to facilitate durable tumor immunity. Oncoimmunology 2018; 8:e1539614. [PMID: 30713799 DOI: 10.1080/2162402x.2018.1539614] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 12/29/2022] Open
Abstract
Metastatic melanoma is a significant clinical problem with a 5-year survival rate of only 15-20%. Recent approval of new immunotherapies and targeted inhibitors have provided much needed options for these patients, in some cases promoting dramatic disease regressions. In particular, antibody-based therapies that block the PD-1/PD-L1 checkpoint inhibitory pathway have achieved an increased overall response rate in metastatic melanoma, yet durable response rates are reported only around 15%. To improve the overall and durable response rates for advanced-stage melanoma, combined targeted and immune-based therapies are under investigation. Here, we investigated how the natural products called schweinfurthins, which have selective anti-proliferative activity against many cancer types, impact anti-(α)PD-1-mediated immunotherapy of murine melanomas. Two different compounds efficiently reduced the growth of human and murine melanoma cells in vitro and induced plasma membrane surface localization of the ER-resident protein calreticulin in B16.F10 melanoma cells, an indicator of immunogenic cell death. In addition, both compounds improved αPD-1-mediated immunotherapy of established tumors in immunocompetent C57BL/6 mice either by delaying tumor progression or resulting in complete tumor regression. Improved immunotherapy was accomplished following only a 5-day course of schweinfurthin, which was associated with initial tumor regression even in the absence of αPD-1. Schweinfurthin-induced tumor regression required an intact immune system as tumors were unaffected in NOD scid gamma (NSG) mice. These results indicate that schweinfurthins improve αPD-1 therapy, leading to enhanced and durable anti-tumor immunity and support the translation of this novel approach to further improve response rates for metastatic melanoma.
Collapse
Affiliation(s)
- Kathleen M Kokolus
- Department of Microbiology & Immunology, Penn State College of Medicine, Hershey, PA, USA
| | - Jeremy S Haley
- Department of Microbiology & Immunology, Penn State College of Medicine, Hershey, PA, USA
| | - Emily J Koubek
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA.,Department of Medicine, Penn State College of Medicine, Hershey, PA, USA
| | - Raghavendra Gowda
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Saketh S Dinavahi
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Arati Sharma
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA.,Penn State Cancer Institute, Hershey, PA, USA
| | - David F Claxton
- Department of Medicine, Penn State College of Medicine, Hershey, PA, USA.,Penn State Cancer Institute, Hershey, PA, USA
| | - Klaus F Helm
- Department of Pathology, Penn State College of Medicine, Hershey, PA, USA.,Department of Dermatology, Penn State College of Medicine, Hershey, PA, USA
| | - Joseph J Drabick
- Department of Medicine, Penn State College of Medicine, Hershey, PA, USA.,Department of Pathology, Penn State College of Medicine, Hershey, PA, USA.,Penn State Melanoma and Skin Cancer Center, Hershey, PA, USA
| | - Gavin P Robertson
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA.,Penn State Cancer Institute, Hershey, PA, USA.,Penn State Melanoma and Skin Cancer Center, Hershey, PA, USA
| | - Jeffrey D Neighbors
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA.,Department of Medicine, Penn State College of Medicine, Hershey, PA, USA.,Penn State Cancer Institute, Hershey, PA, USA
| | - Raymond J Hohl
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA.,Department of Medicine, Penn State College of Medicine, Hershey, PA, USA.,Penn State Cancer Institute, Hershey, PA, USA
| | - Todd D Schell
- Department of Microbiology & Immunology, Penn State College of Medicine, Hershey, PA, USA.,Penn State Cancer Institute, Hershey, PA, USA.,Penn State Melanoma and Skin Cancer Center, Hershey, PA, USA
| |
Collapse
|
2471
|
Katsarelias D, Olofsson Bagge R. ASO Author Reflections: The Future of Isolated Limb Perfusion. Ann Surg Oncol 2018; 25:914-915. [PMID: 30411268 DOI: 10.1245/s10434-018-6968-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Indexed: 11/18/2022]
Affiliation(s)
- Dimitrios Katsarelias
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Roger Olofsson Bagge
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
2472
|
Nan Tie E, Henderson MA, Gyorki DE. Management of in-transit melanoma metastases: a review. ANZ J Surg 2018; 89:647-652. [PMID: 30414233 DOI: 10.1111/ans.14921] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 09/04/2018] [Accepted: 09/25/2018] [Indexed: 02/06/2023]
Abstract
In-transit metastases (ITM) of cutaneous melanoma are locoregional recurrences confined to the superficial lymphatics that occur in 3.4-6.2% of patients diagnosed with melanoma. ITM are a heterogeneous disease that poses a therapeutic dilemma. Patients may have a prolonged disease trajectory involving multiple or repeat treatment modalities for frequent recurrences. The management of ITM has evolved without the development of a standardized protocol. Owing to the variability of the disease course there are few dedicated clinical trials, with a number of key trials in stage III melanoma excluding ITM patients. Thus, there is a paucity of quality data on the efficacy of the treatment modalities available for ITM and even fewer studies directly comparing modalities. At present the mainstay of ITM treatment is surgical resection, with intralesional therapies, isolated limb infusion and radiotherapy utilized as second-line measures. The developing role of targeted therapies and immunotherapy has yet to be explored completely in these patients. This review addresses the evidence base of the efficacy of the various treatment modalities available and those factors that have impacted their clinical uptake.
Collapse
Affiliation(s)
- Emilia Nan Tie
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Michael A Henderson
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Surgery, St Vincent's Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| | - David E Gyorki
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Surgery, St Vincent's Hospital, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
2473
|
The clinical role of the TME in solid cancer. Br J Cancer 2018; 120:45-53. [PMID: 30413828 PMCID: PMC6325164 DOI: 10.1038/s41416-018-0327-z] [Citation(s) in RCA: 461] [Impact Index Per Article: 65.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Revised: 10/02/2018] [Accepted: 10/09/2018] [Indexed: 12/14/2022] Open
Abstract
The highly complex and heterogenous ecosystem of a tumour not only contains malignant cells, but also interacting cells from the host such as endothelial cells, stromal fibroblasts, and a variety of immune cells that control tumour growth and invasion. It is well established that anti-tumour immunity is a critical hurdle that must be overcome for tumours to initiate, grow and spread and that anti-tumour immunity can be modulated using current immunotherapies to achieve meaningful anti-tumour clinical responses. Pioneering studies in melanoma, ovarian and colorectal cancer have demonstrated that certain features of the tumour immune microenvironment (TME)-in particular, the degree of tumour infiltration by cytotoxic T cells-can predict a patient's clinical outcome. More recently, studies in renal cell cancer have highlighted the importance of assessing the phenotype of the infiltrating T cells to predict early relapse. Furthermore, intricate interactions with non-immune cellular players such as endothelial cells and fibroblasts modulate the clinical impact of immune cells in the TME. Here, we review the critical components of the TME in solid tumours and how they shape the immune cell contexture, and we summarise numerous studies evaluating its clinical significance from a prognostic and theranostic perspective.
Collapse
|
2474
|
Joseph RW, Shillington AC, Macahilig C, Diede SJ, Dave V, Harshaw Q, Liu FX. Factors associated with immunotherapy selection in patients with advanced melanoma. Immunotherapy 2018; 10:1361-1369. [PMID: 30407098 DOI: 10.2217/imt-2018-0150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM To explore factors associated with pembrolizumab (PEMBRO) versus ipilimumab + nivolumab (IPI+NIVO) selection in advanced melanoma. MATERIALS & METHODS Total of 12 academic and satellite clinics contributed to this study. Descriptive and logistic regression analyses were conducted to explore associations between clinical characteristics and treatment choice. Results: Total of 400 patients were included: 200 PEMBRO and 200 IPI+NIVO. Patients were significantly more likely to receive PEMBRO versus IPI+NIVO if they had poorer Eastern Cooperative Oncology Group score, 2-4 versus 0-1 (odds ratio [OR]: 6.6; 95% CI: 3.0-14.7), if they were PD-L1 positive (OR: 4.5; 95% CI: 1.9-10.4) or had BRAF wild-type tumor (OR: 2.2; 95% CI: 1.4-3.6). CONCLUSION Patient factors are significantly associated with treatment selection in advanced melanoma. Outcomes comparisons should take this into consideration.
Collapse
Affiliation(s)
- Richard W Joseph
- Division of Hematology/Oncology, Department of Internal Medicine, Mayo Clinic, Jacksonville, FL, USA
| | | | | | - Scott J Diede
- Oncology Clinical Research, Merck & Co., Inc., Kenilworth, NJ, USA
| | | | - Qing Harshaw
- Statistics and Analytics, EPI-Q Inc. Oakbrook, IL, USA
| | - Frank Xiaoqing Liu
- Center for Observational and Real World Evidence, Merck & Co., Inc., Kenilworth, NJ, USA
| |
Collapse
|
2475
|
Xu C, Chen YP, Du XJ, Liu JQ, Huang CL, Chen L, Zhou GQ, Li WF, Mao YP, Hsu C, Liu Q, Lin AH, Tang LL, Sun Y, Ma J. Comparative safety of immune checkpoint inhibitors in cancer: systematic review and network meta-analysis. BMJ 2018; 363:k4226. [PMID: 30409774 PMCID: PMC6222274 DOI: 10.1136/bmj.k4226] [Citation(s) in RCA: 379] [Impact Index Per Article: 54.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/04/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To provide a complete toxicity profile, toxicity spectrum, and a safety ranking of immune checkpoint inhibitor (ICI) drugs for treatment of cancer. DESIGN Systematic review and network meta-analysis. DATA SOURCES Electronic databases (PubMed, Embase, Cochrane Library, and Web of Science) were systematically searched to include relevant studies published in English between January 2007 and February 2018. REVIEW METHODS Only head-to-head phase II and III randomised controlled trials comparing any two or three of the following treatments or different doses of the same ICI drug were included: nivolumab, pembrolizumab, ipilimumab, tremelimumab, atezolizumab, conventional therapy (chemotherapy, targeted therapy, and their combinations), two ICI drugs, or one ICI drug with conventional therapy. Eligible studies must have reported site, organ, or system level data on treatment related adverse events. High quality, single arm trials and placebo controlled trials on ICI drugs were selected to establish a validation group. RESULTS 36 head-to-head phase II and III randomised trials (n=15 370) were included. The general safety of ICI drugs ranked from high to low for all adverse events was as follows: atezolizumab (probability 76%, pooled incidence 66.4%), nivolumab (56%, 71.8%), pembrolizumab (55%, 75.1%), ipilimumab (55%, 86.8%), and tremelimumab (54%, not applicable). The general safety of ICI drugs ranked from high to low for severe or life threatening adverse events was as follows: atezolizumab (49%, 15.1%), nivolumab (46%, 14.1%), pembrolizumab (72%, 19.8%), ipilimumab (51%, 28.6%), and tremelimumab (28%, not applicable). Compared with conventional therapy, treatment-related adverse events for ICI drugs occurred mainly in the skin, endocrine, hepatic, and pulmonary systems. Taking one ICI drug was generally safer than taking two ICI drugs or one ICI drug with conventional therapy. Among the five ICI drugs, atezolizumab had the highest risk of hypothyroidism, nausea, and vomiting. The predominant treatment-related adverse events for pembrolizumab were arthralgia, pneumonitis, and hepatic toxicities. The main treatment-related adverse events for ipilimumab were skin, gastrointestinal, and renal toxicities. Nivolumab had a narrow and mild toxicity spectrum, mainly causing endocrine toxicities. Integrated evidence from the pooled incidences, subgroup, and sensitivity analyses implied that nivolumab is the best option in terms of safety, especially for the treatment of lung cancer. CONCLUSIONS Compared with other ICI drugs used to treat cancer, atezolizumab had the best safety profile in general, and nivolumab had the best safety profile in lung cancer when taking an integrated approach. The safety ranking of treatments based on ICI drugs is modulated by specific treatment-related adverse events. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42017082553.
Collapse
MESH Headings
- Antibodies, Monoclonal/adverse effects
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Clinical Trials, Phase II as Topic
- Clinical Trials, Phase III as Topic
- Drug-Related Side Effects and Adverse Reactions/etiology
- Humans
- Ipilimumab/adverse effects
- Ipilimumab/therapeutic use
- Neoplasms/drug therapy
- Neoplasms/immunology
- Nivolumab/adverse effects
- Nivolumab/therapeutic use
- Randomized Controlled Trials as Topic
Collapse
Affiliation(s)
- Cheng Xu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China
| | - Yu-Pei Chen
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China
| | - Xiao-Jing Du
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China
| | - Jin-Qi Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China
| | - Cheng-Long Huang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China
| | - Lei Chen
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China
| | - Guan-Qun Zhou
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China
| | - Wen-Fei Li
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China
| | - Yan-Ping Mao
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China
| | - Chiun Hsu
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Qing Liu
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ai-Hua Lin
- Department of Medical Statistics and Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou, China
| | - Ling-Long Tang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China
| | - Ying Sun
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China
| | - Jun Ma
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou 510060, China
| |
Collapse
|
2476
|
Smith JS, Nicholson LT, Suwanpradid J, Glenn RA, Knape NM, Alagesan P, Gundry JN, Wehrman TS, Atwater AR, Gunn MD, MacLeod AS, Rajagopal S. Biased agonists of the chemokine receptor CXCR3 differentially control chemotaxis and inflammation. Sci Signal 2018; 11:11/555/eaaq1075. [PMID: 30401786 DOI: 10.1126/scisignal.aaq1075] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The chemokine receptor CXCR3 plays a central role in inflammation by mediating effector/memory T cell migration in various diseases; however, drugs targeting CXCR3 and other chemokine receptors are largely ineffective in treating inflammation. Chemokines, the endogenous peptide ligands of chemokine receptors, can exhibit so-called biased agonism by selectively activating either G protein- or β-arrestin-mediated signaling after receptor binding. Biased agonists might be used as more targeted therapeutics to differentially regulate physiological responses, such as immune cell migration. To test whether CXCR3-mediated physiological responses could be segregated by G protein- and β-arrestin-mediated signaling, we identified and characterized small-molecule biased agonists of the receptor. In a mouse model of T cell-mediated allergic contact hypersensitivity (CHS), topical application of a β-arrestin-biased, but not a G protein-biased, agonist potentiated inflammation. T cell recruitment was increased by the β-arrestin-biased agonist, and biopsies of patients with allergic CHS demonstrated coexpression of CXCR3 and β-arrestin in T cells. In mouse and human T cells, the β-arrestin-biased agonist was the most efficient at stimulating chemotaxis. Analysis of phosphorylated proteins in human lymphocytes showed that β-arrestin-biased signaling activated the kinase Akt, which promoted T cell migration. This study demonstrates that biased agonists of CXCR3 produce distinct physiological effects, suggesting discrete roles for different endogenous CXCR3 ligands and providing evidence that biased signaling can affect the clinical utility of drugs targeting CXCR3 and other chemokine receptors.
Collapse
Affiliation(s)
- Jeffrey S Smith
- Department of Biochemistry, Duke University, Durham, NC 27710, USA.,Department of Medicine, Duke University, Durham, NC 27710, USA
| | | | | | - Rachel A Glenn
- Department of Biochemistry, Duke University, Durham, NC 27710, USA
| | - Nicole M Knape
- Department of Biochemistry, Duke University, Durham, NC 27710, USA
| | - Priya Alagesan
- Department of Biochemistry, Duke University, Durham, NC 27710, USA
| | - Jaimee N Gundry
- Department of Biochemistry, Duke University, Durham, NC 27710, USA
| | | | | | - Michael D Gunn
- Department of Medicine, Duke University, Durham, NC 27710, USA.,Department of Immunology, Duke University, Durham, NC 27710, USA
| | - Amanda S MacLeod
- Department of Dermatology, Duke University, Durham, NC 27710, USA.,Department of Immunology, Duke University, Durham, NC 27710, USA
| | - Sudarshan Rajagopal
- Department of Biochemistry, Duke University, Durham, NC 27710, USA. .,Department of Medicine, Duke University, Durham, NC 27710, USA
| |
Collapse
|
2477
|
Lacey J, Lomax AJ, McNeil C, Marthick M, Levy D, Kao S, Nielsen T, Dhillon HM. A supportive care intervention for people with metastatic melanoma being treated with immunotherapy: a pilot study assessing feasibility, perceived benefit, and acceptability. Support Care Cancer 2018; 27:1497-1507. [PMID: 30392112 DOI: 10.1007/s00520-018-4524-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/19/2018] [Indexed: 01/03/2023]
Abstract
INTRODUCTION Increasing numbers of metastatic melanoma (MM) patients are receiving immunotherapy treatment, including pembrolizumab, and the impact on their well-being is underexplored. OBJECTIVES To assess the feasibility of a multimodal supportive care program to MM patients being treated with pembrolizumab. METHODS This pre-post-test feasibility cohort study recruited MM participants treated with pembrolizumab: (i) supportive care intervention with usual care and (ii) usual care. The intervention comprised comprehensive medical assessment by supportive care physician (SCP), exercise physiologist (EP), and dietitian then a tailored supportive care program. Programs included exercise, dietary advice, non-invasive complementary therapies, and psychology consultation. Outcome measures included adherence, patient-reported symptoms, anxiety and depression, and toxicity. Descriptive data are reported. RESULTS We recruited 28 participants: 13 intervention and 15 control; three did not complete the study. Most were male, with median age 66 (range 42-85) years. All intervention participants completed baseline assessments with SCP, EP, and dietitian. Two missed follow-up with EP or dietitian. Symptoms most troubling at baseline were as follows: fatigue (n = 6), sleep (n = 6), general aches and pains (n = 5), and memory (n = 4). All intervention participants were prescribed 16 exercise sessions; 8 (50%) completed all; overall exercise adherence was 85%. Integrative therapies were accessed by 85% (11) participants. Immunotherapy-related adverse event rates were low and SCP consultation identified symptoms not captured by CTCAE 4.0. CONCLUSIONS A holistic supportive care intervention tailored to individual needs is feasible. The symptom burden in MM patients was low. Further investigation of the intervention is warranted, focused on populations with higher symptom burden to improve outcomes.
Collapse
Affiliation(s)
- Judith Lacey
- Chris O'Brien Lifehouse Comprehensive Cancer Centre, Missenden Rd, Camperdown, NSW, Australia.
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia.
| | - Anna J Lomax
- Chris O'Brien Lifehouse Comprehensive Cancer Centre, Missenden Rd, Camperdown, NSW, Australia
| | - Catriona McNeil
- Chris O'Brien Lifehouse Comprehensive Cancer Centre, Missenden Rd, Camperdown, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Michael Marthick
- Chris O'Brien Lifehouse Comprehensive Cancer Centre, Missenden Rd, Camperdown, NSW, Australia
| | - David Levy
- Chris O'Brien Lifehouse Comprehensive Cancer Centre, Missenden Rd, Camperdown, NSW, Australia
- Centre for Medical Psychology and Evidence-Based Decision-Making, School of Psychology, University of Sydney, Sydney, NSW, Australia
| | - Steven Kao
- Chris O'Brien Lifehouse Comprehensive Cancer Centre, Missenden Rd, Camperdown, NSW, Australia
- Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Theresa Nielsen
- Chris O'Brien Lifehouse Comprehensive Cancer Centre, Missenden Rd, Camperdown, NSW, Australia
| | - Haryana M Dhillon
- Centre for Medical Psychology and Evidence-Based Decision-Making, School of Psychology, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
2478
|
Kang A, Zhao D, Yeh JJ, Lee DJ. Updates on Immunotherapy for the Treatment of Skin Cancer. CURRENT DERMATOLOGY REPORTS 2018. [DOI: 10.1007/s13671-018-0246-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
2479
|
Ascierto PA, Flaherty K, Goff S. Emerging Strategies in Systemic Therapy for the Treatment of Melanoma. Am Soc Clin Oncol Educ Book 2018; 38:751-758. [PMID: 30231371 DOI: 10.1200/edbk_199047] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Recent years have seen major improvements in survival of patients with advanced melanoma with the advent of various novel systemic immunotherapies and targeted therapies. As our understanding of these agents and their various mechanisms of action improves, even more impressive outcomes are being achieved through use of various combination strategies, including the combining of different immunotherapies with one another as well as with other modalities. However, despite the improved outcomes that have been achieved in advanced melanoma, responses to treatment are heterogeneous and may not always be durable. Additional advances in therapy are required, and several emerging strategies are a focus of interest. These include the investigation of several new immunotherapy and/or targeted therapy combinations, such as checkpoint inhibitors (anti-PD-1/anti-CTLA-4) with other immunotherapies (e.g., indoleamine 2,3 dioxygenase [IDO] inhibitors, antilymphocyte activation 3 [anti-LAG-3], histone deacetylase [HDAC] inhibitors, Toll-like receptor 9 [TLR-9] agonists, antiglucocorticoid-induced tumor necrosis factor receptor [anti-GITR], pegylated interleukin-2 [IL-2]), combined targeted therapies (e.g., MEK and CDK4/6 coinhibition), and combined immunotherapy and targeted therapy (e.g., the triplet combination of BRAF/MEK inhibition with anti-PD-1s). The identification of novel therapeutic targets in the MAP kinase pathway also offers opportunities to improve outcomes by overcoming de novo and acquired resistance to BRAF/MEK inhibition (e.g., the development of ERK inhibitors). In addition, adoptive cell transfer, the infusion of large numbers of activated autologous lymphocytes, may have a potential role in patients whose disease has progressed after immunotherapy. Taken together, these new approaches offer further potential to increase systemic treatment options and improve long-term outcomes for patients with advanced melanoma.
Collapse
Affiliation(s)
- Paolo A Ascierto
- From the Istituto Nazionale Tumori "Fondazione G. Pascale," Naples, Italy; Massachusetts General Hospital Cancer Center, Boston, MA; Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Keith Flaherty
- From the Istituto Nazionale Tumori "Fondazione G. Pascale," Naples, Italy; Massachusetts General Hospital Cancer Center, Boston, MA; Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Stephanie Goff
- From the Istituto Nazionale Tumori "Fondazione G. Pascale," Naples, Italy; Massachusetts General Hospital Cancer Center, Boston, MA; Center for Cancer Research, National Cancer Institute, Bethesda, MD
| |
Collapse
|
2480
|
Park JS, Kim JH. Role of non-classical T cells in skin immunity. Mol Immunol 2018; 103:286-292. [DOI: 10.1016/j.molimm.2018.09.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 09/14/2018] [Accepted: 09/29/2018] [Indexed: 12/30/2022]
|
2481
|
Amaria RN, Reddy SM, Tawbi HA, Davies MA, Ross MI, Glitza IC, Cormier JN, Lewis C, Hwu WJ, Hanna E, Diab A, Wong MK, Royal R, Gross N, Weber R, Lai SY, Ehlers R, Blando J, Milton DR, Woodman S, Kageyama R, Wells DK, Hwu P, Patel SP, Lucci A, Hessel A, Lee JE, Gershenwald J, Simpson L, Burton EM, Posada L, Haydu L, Wang L, Zhang S, Lazar AJ, Hudgens CW, Gopalakrishnan V, Reuben A, Andrews MC, Spencer CN, Prieto V, Sharma P, Allison J, Tetzlaff MT, Wargo JA. Neoadjuvant immune checkpoint blockade in high-risk resectable melanoma. Nat Med 2018; 24:1649-1654. [PMID: 30297909 PMCID: PMC6481682 DOI: 10.1038/s41591-018-0197-1] [Citation(s) in RCA: 609] [Impact Index Per Article: 87.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 08/21/2018] [Indexed: 12/17/2022]
Abstract
Preclinical studies suggest that treatment with neoadjuvant immune checkpoint blockade is associated with enhanced survival and antigen-specific T cell responses compared with adjuvant treatment1; however, optimal regimens have not been defined. Here we report results from a randomized phase 2 study of neoadjuvant nivolumab versus combined ipilimumab with nivolumab in 23 patients with high-risk resectable melanoma ( NCT02519322 ). RECIST overall response rates (ORR), pathologic complete response rates (pCR), treatment-related adverse events (trAEs) and immune correlates of response were assessed. Treatment with combined ipilimumab and nivolumab yielded high response rates (RECIST ORR 73%, pCR 45%) but substantial toxicity (73% grade 3 trAEs), whereas treatment with nivolumab monotherapy yielded modest responses (ORR 25%, pCR 25%) and low toxicity (8% grade 3 trAEs). Immune correlates of response were identified, demonstrating higher lymphoid infiltrates in responders to both therapies and a more clonal and diverse T cell infiltrate in responders to nivolumab monotherapy. These results describe the feasibility of neoadjuvant immune checkpoint blockade in melanoma and emphasize the need for additional studies to optimize treatment regimens and to validate putative biomarkers.
Collapse
Affiliation(s)
- Rodabe N. Amaria
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Sangeetha M. Reddy
- Department of Breast Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Hussein A. Tawbi
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael A. Davies
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Merrick I. Ross
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Isabella C. Glitza
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Janice N. Cormier
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Carol Lewis
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Wen-Jen Hwu
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Ehab Hanna
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Adi Diab
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael K. Wong
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Richard Royal
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Neil Gross
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Randal Weber
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Stephen Y. Lai
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Richard Ehlers
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jorge Blando
- Department of Immunology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Denái R. Milton
- Department of Biostatistics, MD Anderson Cancer Center, Houston, Texas, USA
| | - Scott Woodman
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Robin Kageyama
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Danny K. Wells
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | - Patrick Hwu
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Sapna P. Patel
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Anthony Lucci
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Amy Hessel
- Department of Head and Neck Surgery, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeffrey E. Lee
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeffrey Gershenwald
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Lauren Simpson
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Elizabeth M. Burton
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Liberty Posada
- Department of Melanoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Lauren Haydu
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Linghua Wang
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| | - Shaojun Zhang
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | | | - Alexandre Reuben
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Miles C. Andrews
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Victor Prieto
- Department of Immunology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Padmanee Sharma
- Department of Immunology, MD Anderson Cancer Center, Houston, Texas, USA
- Department of Genitourinary Cancers, MD Anderson Cancer Center, Houston, Texas, USA
| | - James Allison
- Department of Immunology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Michael T. Tetzlaff
- Department of Pathology, MD Anderson Cancer Center, Houston, Texas, USA
- Department of Translational and Molecular Pathology, MD Anderson Cancer Center, Houston, Texas, USA
| | - Jennifer A. Wargo
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, Texas, USA
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
2482
|
Buerki RA, Chheda ZS, Okada H. Immunotherapy of Primary Brain Tumors: Facts and Hopes. Clin Cancer Res 2018; 24:5198-5205. [PMID: 29871908 PMCID: PMC6214775 DOI: 10.1158/1078-0432.ccr-17-2769] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 12/28/2022]
Abstract
The field of cancer immunotherapy has made exciting progress for some cancer types in recent years. However, recent failures of late-phase clinical trials evaluating checkpoint blockade in patients with glioblastoma (GBM) represent continued challenges for brain cancer immunotherapy. This is likely due to multiple factors including but not limited to marked genetic and antigenic heterogeneity, relatively low mutational loads, and paucity of GBM-infiltrating T cells. We review recent and ongoing studies targeting the checkpoint molecules as monotherapy or in combination with other modalities, and discuss the mechanisms underlying the unresponsiveness of GBM to single-modality immunotherapy approaches. We also discuss other novel immunotherapy approaches that may promote T-cell responses and overcome the "cold tumor" status of GBM, including oncolytic viruses and adoptive T-cell therapy. Clin Cancer Res; 24(21); 5198-205. ©2018 AACR.
Collapse
Affiliation(s)
- Robin A Buerki
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Zinal S Chheda
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Hideho Okada
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California.
- The Parker Institute for Cancer Immunotherapy, San Francisco, California
- Cancer Immunotherapy Program, University of California, San Francisco, San Francisco, California
| |
Collapse
|
2483
|
Passaro C, Somma SD, Malfitano AM, Portella G. Oncolytic virotherapy for anaplastic and poorly differentiated thyroid cancer: a promise or a clinical reality? INTERNATIONAL JOURNAL OF ENDOCRINE ONCOLOGY 2018. [DOI: 10.2217/ije-2017-0028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Oncolytic viruses (OVs) selectively infect and lyse cancer cells. A direct lytic effect of OVs has been theorized in the initial studies; however, the antineoplastic effect of OVs is also due to the induction of an immune response against cancer cells. Anaplastic thyroid cancer is one of the most aggressive human malignancies with a short survival time of about 6–12 months from the diagnosis. The lack of effective therapies has prompted to investigate the efficacy of OVs in anaplastic thyroid carcinoma. Different OVs have been tested in preclinical studies, either as single agents or in combinatorial treatments. In this review, the results of these studies are summarized and future perspective discussed.
Collapse
Affiliation(s)
- Carmela Passaro
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli Federico II, Napoli, Italia
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sarah Di Somma
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli Federico II, Napoli, Italia
| | - Anna Maria Malfitano
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli Federico II, Napoli, Italia
| | - Giuseppe Portella
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli Federico II, Napoli, Italia
| |
Collapse
|
2484
|
Soularue E, Lepage P, Colombel JF, Coutzac C, Faleck D, Marthey L, Collins M, Chaput N, Robert C, Carbonnel F. Enterocolitis due to immune checkpoint inhibitors: a systematic review. Gut 2018; 67:2056-2067. [PMID: 30131322 DOI: 10.1136/gutjnl-2018-316948] [Citation(s) in RCA: 178] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/11/2018] [Accepted: 07/19/2018] [Indexed: 12/14/2022]
Abstract
Immune checkpoint inhibitors targeting cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) and programmed death-1 (PD-1)/ligand are increasingly used to treat several types of cancer. These drugs enhance antitumour T-cell activity and therefore induce immune-related adverse effects (irAE), of which gastrointestinal (GI) irAE are among the most frequent and severe. This systematic literature review summarises the clinical manifestations, management and pathophysiology of GI irAE due to immune checkpoint inhibitors. GI irAE induced by anti-CTLA-4 are frequent, potentially severe and resemble IBD, whereas those induced by PD-1 blockade seem to be less frequent and clinically more diverse. Baseline symbiotic gut microbiota is associated with an enhanced antitumour response to immune checkpoint inhibitors and an increased susceptibility to developing enterocolitis, in patients treated with anti-CTLA-4. These findings open new perspectives for possible manipulation of the gut microbiota in order to better identify responders to immune checkpoint inhibitors and to increase their efficacy and safety.
Collapse
Affiliation(s)
- Emilie Soularue
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique- Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France.,Faculté de Médecine, University Paris-Saclay, Le Kremlin Bicêtre, France
| | - Patricia Lepage
- Micalis Institute, INRA, AgroParisTech, University Paris-Saclay, Jouy-en- Josas, France
| | - Jean Frederic Colombel
- Helmsley Inflammatory Bowel Disease Center, Icahn Medical School of Medicine at Mount Sinai, New York, USA
| | - Clelia Coutzac
- Laboratory of Immunomonitoring in Oncology and CNRS-UMS 3655 and INSERM-US23, Villejuif, France
| | - David Faleck
- Helmsley Inflammatory Bowel Disease Center, Icahn Medical School of Medicine at Mount Sinai, New York, USA
| | - Lysiane Marthey
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique- Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France
| | - Michael Collins
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique- Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France.,Faculté de Médecine, University Paris-Saclay, Le Kremlin Bicêtre, France
| | - Nathalie Chaput
- Laboratory of Immunomonitoring in Oncology and CNRS-UMS 3655 and INSERM-US23, Villejuif, France.,Faculté de Pharmacie, University Paris-Saclay, Chatenay-Malabry, France
| | - Caroline Robert
- Faculté de Médecine, University Paris-Saclay, Le Kremlin Bicêtre, France.,Départment of Medecine, Dermatology Unit, Villejuif, France
| | - Franck Carbonnel
- Department of Gastroenterology, Kremlin Bicêtre Hospital, Assistance Publique- Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France.,Faculté de Médecine, University Paris-Saclay, Le Kremlin Bicêtre, France
| |
Collapse
|
2485
|
Patel RB, Baniel CC, Sriramaneni RN, Bradley K, Markovina S, Morris ZS. Combining brachytherapy and immunotherapy to achieve in situ tumor vaccination: A review of cooperative mechanisms and clinical opportunities. Brachytherapy 2018; 17:995-1003. [PMID: 30078541 PMCID: PMC8292980 DOI: 10.1016/j.brachy.2018.07.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 07/06/2018] [Indexed: 12/22/2022]
Abstract
As immunotherapies continue to emerge as a standard component of treatment for a variety of cancers, the imperative for testing these in combination with other standard cancer therapies grows. Radiation therapy may be a particularly well-suited partner for many immunotherapies. By modulating immune tolerance and functional immunogenicity at a targeted tumor site, radiation therapy may serve as a method of in situ tumor vaccination. In situ tumor vaccination is a therapeutic strategy that seeks to convert a patient's own tumor into a nidus for enhanced presentation of tumor-specific antigens in a way that will stimulate and diversify an antitumor T cell response. The mechanisms whereby radiation may impact immunotherapy are diverse and include its capacity to simultaneously elicit local inflammation, temporary local depletion of suppressive lymphocyte lineages, enhanced tumor cell susceptibility to immune response, and immunogenic tumor cell death. Emerging data suggest that each of these mechanisms may display a distinct dose-response profile, making it challenging to maximize each of these effects using external beam radiation. Conversely, the highly heterogenous and conformal dose distribution achieved with brachytherapy may be optimal for enhancing the immunogenic capacity of radiation at a tumor site while minimizing off-target antagonistic effects on peripheral immune cells. Here, we review the immunogenic effects of radiation, summarize the clinical rationale and data supporting the use of radiation together with immunotherapies, and discuss the rationale and urgent need for further preclinical and clinical investigation specifically of brachytherapy in combination with immunotherapies. Harnessing these immunomodulatory effects of brachytherapy may offer solutions to overcome obstacles to the efficacy of immunotherapies in immunologically "cold" tumors while potentiating greater response in the context of immunologically "hot" tumors.
Collapse
Affiliation(s)
- Ravi B Patel
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Claire C Baniel
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Raghava N Sriramaneni
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Kristin Bradley
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Stephanie Markovina
- Department of Radiation Oncology, Washington University in St Louis, St Louis, MO
| | - Zachary S Morris
- Department of Human Oncology, University of Wisconsin School of Medicine and Public Health, Madison, WI.
| |
Collapse
|
2486
|
Indini A, Brecht I, Del Vecchio M, Sultan I, Signoroni S, Ferrari A. Cutaneous melanoma in adolescents and young adults. Pediatr Blood Cancer 2018; 65:e27292. [PMID: 29968969 DOI: 10.1002/pbc.27292] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 05/21/2018] [Accepted: 05/29/2018] [Indexed: 12/24/2022]
Abstract
Cutaneous melanoma is rare in children, but has greater incidence in adolescents and young adults (AYAs). Diagnosis may be challenging due to its rarity in these age groups. Few studies have specifically addressed the topic of AYA melanoma. Though young-age melanoma may have particular biological characteristics, available data suggest that its clinical history is similar to that of adults. However, advances in treatment of adult melanoma have not been reflected in the treatment of AYAs. There is no standard treatment, and access to clinical trials is difficult for AYAs. Further efforts are needed to overcome these issues by improving cooperation with experts on adult melanoma.
Collapse
Affiliation(s)
- Alice Indini
- Melanoma Medical Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Ines Brecht
- Department of Pediatric Hematology and Oncology, University of Tubingen, Tubingen, Germany
| | - Michele Del Vecchio
- Melanoma Medical Oncology Unit, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Iyad Sultan
- Department of Pediatric Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Stefano Signoroni
- Unit of Hereditary Digestive Tract Tumours, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Andrea Ferrari
- Pediatric Oncology Unit, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| |
Collapse
|
2487
|
McKean MA, Amaria RN. Multidisciplinary treatment strategies in high-risk resectable melanoma: Role of adjuvant and neoadjuvant therapy. Cancer Treat Rev 2018; 70:144-153. [DOI: 10.1016/j.ctrv.2018.08.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 08/27/2018] [Accepted: 08/28/2018] [Indexed: 12/17/2022]
|
2488
|
Ascierto PA, Marabelle A. How do immune checkpoint-targeted antibodies work? The need for improved pharmacokinetic evaluation in early phase studies. Ann Oncol 2018; 29:2157-2160. [PMID: 30307539 DOI: 10.1093/annonc/mdy420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- P A Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italic.
| | - A Marabelle
- Département d'Innovation Thérapeutique et d'Essais Précoces, Gustave Roussy, Université Paris-Saclay, Villejuif; INSERM U1015, Villejuif, France
| |
Collapse
|
2489
|
Tio M, Rai R, Ezeoke OM, McQuade JL, Zimmer L, Khoo C, Park JJ, Spain L, Turajlic S, Ardolino L, Yip D, Goldinger SM, Cohen JV, Millward M, Atkinson V, Kane AY, Ascierto PA, Garbe C, Gutzmer R, Johnson DB, Rizvi HA, Joshua AM, Hellmann MD, Long GV, Menzies AM. Anti-PD-1/PD-L1 immunotherapy in patients with solid organ transplant, HIV or hepatitis B/C infection. Eur J Cancer 2018; 104:137-144. [PMID: 30347289 PMCID: PMC10176037 DOI: 10.1016/j.ejca.2018.09.017] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 09/20/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Anti-programmed cell death protein 1/programmed death ligand 1 (PD-1/PD-L1) immunotherapy is now routinely used to treat several cancers. Clinical trials have excluded several populations, including patients with solid organ transplant, HIV infection and hepatitis B/C infection. We examined the safety outcomes of these populations treated with anti-PD-1/PD-L1 treatment in a multicentre retrospective study. METHODS Patients from 16 centres with advanced cancer and solid organ transplant, HIV infection or hepatitis B/C infection were included. Demographic, tumour, treatment, toxicity and outcome data were recorded. RESULTS Forty-six patients were included for analysis, with a median age of 60 years, and the majority of patients diagnosed with melanoma (72%). Among six patients with solid organ transplants, two graft rejections occurred, with one resulting in death, whereas two patients achieved partial responses. There were four responses in 12 patients with HIV infection. In 14 patients with hepatitis B, there were three responses, and similarly, there were three responses in 14 patients with hepatitis C. There was no unexpected toxicity in any viral infection group or an increase in viral load. CONCLUSION Patients with HIV or hepatitis B/C infections treated with anti-PD-1/PD-L1 immunotherapy may respond to treatment without increased toxicity. Given the risk of graft rejection in solid organ transplant patients and also the potential for response, the role of anti-PD-1/PD-L1 immunotherapy needs to be carefully considered.
Collapse
Affiliation(s)
- Martin Tio
- Melanoma Institute Australia, Sydney, Australia.
| | - Rajat Rai
- Melanoma Institute Australia, Sydney, Australia
| | | | | | - Lisa Zimmer
- University of Duisburg-Essen Hospital, Heidelberg, Germany
| | - Chloe Khoo
- Peter MacCallum Cancer Centre, Melbourne, Australia
| | - John J Park
- Crown Princess Mary Cancer Centre, Sydney, Australia; Westmead Hospital, Sydney, Australia
| | - Lavinia Spain
- Skin and Renal Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - Samra Turajlic
- Skin and Renal Unit, The Royal Marsden NHS Foundation Trust, London, UK; Francis Crick Institute, London, UK
| | | | - Desmond Yip
- The Canberra Hospital, Canberra, Australia; ANU Medical School, Australian National University, Canberra, Australia
| | | | | | | | | | - Alisa Y Kane
- Liverpool Hospital, Sydney, Australia; Garvan Institute, Sydney, Australia
| | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | | | | | | | - Hira A Rizvi
- Memorial Sloan Kettering Cancer Center, New York, USA
| | - Anthony M Joshua
- Melanoma Institute Australia, Sydney, Australia; St Vincents Hospital, Sydney, Australia; University of New South Wales, Sydney, Australia
| | | | - Georgina V Long
- Melanoma Institute Australia, Sydney, Australia; The University of Sydney, Sydney, Australia; Royal North Shore Hospital, Sydney, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, Sydney, Australia; The University of Sydney, Sydney, Australia; Royal North Shore Hospital, Sydney, Australia
| |
Collapse
|
2490
|
Kartolo A, Sattar J, Sahai V, Baetz T, Lakoff JM. Predictors of immunotherapy-induced immune-related adverse events. ACTA ACUST UNITED AC 2018; 25:e403-e410. [PMID: 30464691 DOI: 10.3747/co.25.4047] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Purpose We aimed to elucidate predictive factors for the development of immune-related adverse events (iraes) in patients receiving immunotherapies for the management of advanced solid cancers. Methods This retrospective study involved all patients with histologically confirmed metastatic or inoperable melanoma, non-small-cell lung cancer, or renal cell carcinoma receiving immunotherapy at the Cancer Centre of Southeastern Ontario. The type and severity of iraes, as well as potential protective and exacerbating factors, were collected from patient charts. Results The study included 78 patients receiving ipilimumab (32%), nivolumab (33%), or pembrolizumab (35%). Melanoma, non-small-cell lung cancer, and renal cell carcinoma accounted for 70%, 22%, and 8% of the cancers in the study population. In 41 patients (53%) iraes developed, with multiple iraes developing in 12 patients (15%). In most patients (70%), the iraes were of severity grade 1 or 2. Female sex [adjusted odds ratio (oradj): 0.094; 95% confidence interval (ci): 0.021 to 0.415; p = 0.002] and corticosteroid use before immunotherapy (oradj: 0.143; 95% ci: 0.036 to 0.562; p = 0.005) were found to be associated with a protective effect against iraes. In contrast, a history of autoimmune disease (oradj: 9.55; 95% ci: 1.34 to 68.22; p = 0.025), use of ctla-4 inhibitors (oradj: 6.25; 95% ci: 1.61 to 24.25; p = 0.008), and poor kidney function of grade 3 or greater (oradj: 10.66; 95% ci: 2.41 to 47.12; p = 0.025) were associated with a higher risk of developing iraes. A Hosmer-Lemeshow goodness-of-fit test demonstrated that the logistic regression model was effective at predicting the development of iraes (chi-square: 1.596; df = 7; p = 0.979). Conclusions Our study highlights several factors that affect the development of iraes in patients receiving immunotherapy. Although future studies are needed to validate the resulting model, findings from the study can help to guide risk stratification, monitoring, and management of iraes in patients given immunotherapy for advanced cancer.
Collapse
Affiliation(s)
- A Kartolo
- Department of Medicine, Queen's University, Kingston, ON
| | - J Sattar
- Department of Medicine, Queen's University, Kingston, ON
| | - V Sahai
- Hotel Dieu Hospital, Kingston, ON
| | - T Baetz
- Department of Medicine, Queen's University, Kingston, ON.,Cancer Centre of Southeastern Ontario, Kingston, ON
| | - J M Lakoff
- Department of Medicine, Queen's University, Kingston, ON.,Hotel Dieu Hospital, Kingston, ON.,Department of Endocrinology, Queen's University, Kingston, ON
| |
Collapse
|
2491
|
Wang DY, Mooradian MJ, Kim D, Shah NJ, Fenton SE, Conry RM, Mehta R, Silk AW, Zhou A, Compton ML, Al-Rohil RN, Lee S, Voorhees AL, Ha L, McKee S, Norrell JT, Mehnert J, Puzanov I, Sosman JA, Chandra S, Gibney GT, Rapisuwon S, Eroglu Z, Sullivan R, Johnson DB. Clinical characterization of colitis arising from anti-PD-1 based therapy. Oncoimmunology 2018; 8:e1524695. [PMID: 30546965 PMCID: PMC6287774 DOI: 10.1080/2162402x.2018.1524695] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 09/10/2018] [Accepted: 09/12/2018] [Indexed: 12/17/2022] Open
Abstract
Colitis is a frequent, clinically-significant immune-related adverse event caused by anti-programmed death-1 (PD-1). The clinical features, timing, and management of colitis with anti-PD-1-based regimens are not well-characterized. Patients with advanced melanoma that received either anti-PD-1 monotherapy ("monotherapy") or combined with ipilimumab ("combination therapy") were screened from 8 academic medical centers, to identify those with clinically-relevant colitis (colitis requiring systemic steroids). Of 1261 patients who received anti-PD-1-based therapy, 109 experienced colitis. The incidence was 3.2% (30/937) and 24.4% (79/324) in the monotherapy and combination therapy cohorts, respectively. Patients with colitis from combination therapy had significantly earlier symptom onset (7.2 weeks vs 25.4 weeks, p < 0.0001), received higher steroid doses (median prednisone equivalent 1.5 mg/kg vs 1.0 mg/kg, p = 0.0015) and experienced longer steroid tapers (median 6.0 vs 4.0 weeks, p = 0.0065) compared to monotherapy. Infliximab use and steroid-dose escalation occurred more frequently in the combination therapy cohort compared to monotherapy. Nearly all patients had resolution of their symptoms although one patient died from complications. Anti-PD-1 associated colitis has a variable clinical presentation, and is more frequent and severe when associated with combination therapy. This variability in checkpoint-inhibitor associated colitis suggests that further optimization of treatment algorithms is needed.
Collapse
Affiliation(s)
- Daniel Y Wang
- Department of Medicine, Vanderbilt University, Nashville, TN, USA,CONTACT Douglas B. Johnson Vanderbilt University Medical Center, 2220 Pierce Ave, 777 Preston Research Bldg. Nashville TN 37232
| | | | - DaeWon Kim
- Department of Medicine, Moffitt Cancer Center, Tampa, FL, USA
| | - Neil J Shah
- Department of Medicine, Georgetown University-Lombardi Cancer Center, Washington, DC, USA
| | - Sarah E Fenton
- Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Robert M Conry
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Rutika Mehta
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, IL, USA
| | - Ann W. Silk
- Department of Medicine, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Alice Zhou
- Department of Medicine, Vanderbilt University, Nashville, TN, USA
| | - Margaret L Compton
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, USA
| | - Rami N Al-Rohil
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University, Nashville, TN, USA
| | - Sunyoung Lee
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, IL, USA
| | | | - Lisa Ha
- Department of Medicine, Northwestern University, Chicago, IL, USA
| | - Svetlana McKee
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jacqueline T Norrell
- Department of Medicine, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Janice Mehnert
- Department of Medicine, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Igor Puzanov
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, IL, USA
| | - Jeffrey A Sosman
- Department of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Geoffrey T Gibney
- Department of Medicine, Georgetown University-Lombardi Cancer Center, Washington, DC, USA
| | - Suthee Rapisuwon
- Department of Medicine, Georgetown University-Lombardi Cancer Center, Washington, DC, USA
| | - Zeynep Eroglu
- Department of Medicine, Moffitt Cancer Center, Tampa, FL, USA
| | - Ryan Sullivan
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | | |
Collapse
|
2492
|
Retrospective Side Effect Profiling of the Metastatic Melanoma Combination Therapy Ipilimumab-Nivolumab Using Adverse Event Data. Diagnostics (Basel) 2018; 8:diagnostics8040076. [PMID: 30384507 PMCID: PMC6316083 DOI: 10.3390/diagnostics8040076] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 10/29/2018] [Accepted: 10/30/2018] [Indexed: 12/15/2022] Open
Abstract
Recent studies suggest that combining nivolumab with ipilimumab is a more effective treatment for melanoma patients, compared to using ipilimumab or nivolumab alone. However, treatment with these immunotherapeutic agents is frequently associated with increased risk of toxicity, and (auto-) immune-related adverse events. The precise pathophysiologic mechanisms of these events are not yet clear, and evidence from clinical trials and translational studies remains limited. Our retrospective analysis of ~7700 metastatic melanoma patients treated with ipilimumab and/or nivolumab from the FDA Adverse Event Reporting System (FAERS) demonstrates that the identified immune-related reactions are specific to ipilimumab and/or nivolumab, and that when the two agents are administered together, their safety profile combines reactions from each drug alone. While more prospective studies are needed to characterize the safety of ipilimumab and nivolumab, the present work constitutes perhaps the first effort to examine the safety of these drugs and their combination based on computational evidence from real world post marketing data.
Collapse
|
2493
|
Lee H, Quek C, Silva I, Tasker A, Batten M, Rizos H, Lim SY, Nur Gide T, Shang P, Attrill GH, Madore J, Edwards J, Carlino MS, Guminski A, Saw RPM, Thompson JF, Ferguson PM, Palendira U, Menzies AM, Long GV, Scolyer RA, Wilmott JS. Integrated molecular and immunophenotypic analysis of NK cells in anti-PD-1 treated metastatic melanoma patients. Oncoimmunology 2018; 8:e1537581. [PMID: 30713793 DOI: 10.1080/2162402x.2018.1537581] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 10/11/2018] [Accepted: 10/13/2018] [Indexed: 10/28/2022] Open
Abstract
Purpose: Anti-PD-1 therapy has revolutionized the treatment and improved the survival of stage IV melanoma patients. However, almost half of the patients fail to respond due to immune evasive mechanism. A known mechanism is the downregulation of major histocompatibility complex (MHC) class I expression, which prevents T cell recognition of the tumor. This study determined the relationship between natural killer (NK) cell numbers and clinical response to anti-PD-1 therapy in metastatic melanoma. Experimental Design: Twenty-five anti-PD-1 treated metastatic melanoma patients were categorized into responders (complete response (CR)/partial response (PR)/stable disease (SD) ≥ 6 mo, n = 13) and non-responders (SD < 6 days/progressive disease (PD), n = 12) based on RECIST response. Whole transcriptome sequencing and multiplex immunofluorescent staining were performed on pre-treatment and on a subset of early during treatment tumor samples. Spatial distribution analysis was performed on multiplex immunofluorescent images to determine the proximity of NK cells to tumor cells. Flow cytometry was used to confirm NK phenotypes in lymph node metastases of treatment naïve melanoma patients (n = 5). Cytotoxic assay was performed using NK cells treated with anti-PD-1 or with isotype control and co-cultured with 3 different melanoma cell lines and with K562 cells (leukemia cell line). Results: Differential expression analysis identified nine upregulated NK cell specific genes (adjusted p < 0.05) in responding (n = 11) versus non-responding patients (n = 10). Immunofluorescent staining of biopsies confirmed a significantly higher density of intra- and peri-tumoral CD16+ and granzyme B + NK cells in responding patients (p < 0.05). Interestingly, NK cells were in closer proximity to tumor cells in responding PD-1 treated patients compared to non-responding patients. Patients who responded to anti-PD-1 therapy, despite MHC class I loss had higher NK cell densities than patients with low MHC class I expression. Lastly, functional assays demonstrated PD-1 blockade induces an increase in NK cells' cytotoxicity. Conclusions: A higher density of tumoral NK cells is associated with response to anti-PD-1 therapy. NK cells may play an important role in mediating response to anti-PD-1 therapy, including in a subset of tumors downregulating MHC class I expression.
Collapse
Affiliation(s)
- Hansol Lee
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,The University of Sydney Central Clinical School, Sydney, NSW, Australia
| | - Camelia Quek
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Ines Silva
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Annie Tasker
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Marcel Batten
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,The University of Sydney Central Clinical School, Sydney, NSW, Australia
| | - Helen Rizos
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Su Yin Lim
- Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Tuba Nur Gide
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,The University of Sydney Central Clinical School, Sydney, NSW, Australia
| | - Ping Shang
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Grace H Attrill
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,The University of Sydney Central Clinical School, Sydney, NSW, Australia
| | - Jason Madore
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,The University of Sydney Central Clinical School, Sydney, NSW, Australia
| | - Jarem Edwards
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,The University of Sydney Central Clinical School, Sydney, NSW, Australia
| | - Matteo S Carlino
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,Crown Princess Mary Cancer Centre, Westmead and Blacktown Hospitals, Sydney, NSW, Australia
| | - Alexander Guminski
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Robyn P M Saw
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - John F Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Peter M Ferguson
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Umaimainthan Palendira
- Discipline of Infectious Diseases and Immunology, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Georgina V Long
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,The University of Sydney Central Clinical School, Sydney, NSW, Australia.,Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - James S Wilmott
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia.,The University of Sydney Central Clinical School, Sydney, NSW, Australia
| |
Collapse
|
2494
|
De Santis F, Del Vecchio M, Castagnoli L, De Braud F, Di Cosimo S, Franceschini D, Fucà G, Hiscott J, Malmberg KJ, McGranahan N, Pietrantonio F, Rivoltini L, Sangaletti S, Tagliabue E, Tripodo C, Vernieri C, Zitvogel L, Pupa SM, Di Nicola M. Innovative therapy, monoclonal antibodies, and beyond: Highlights from the eighth annual meeting. Cytokine Growth Factor Rev 2018; 44:1-10. [PMID: 30393044 DOI: 10.1016/j.cytogfr.2018.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The eighth annual conference of "Innovative therapy, monoclonal antibodies, and beyond" was held in Milan on Jan. 26, 2018, and hosted by Fondazione IRCCS-Istituto Nazionale dei Tumori (Fondazione IRCCS INT). The conference was divided into two main scientific sessions, of i) pre-clinical assays and novel biotargets, and ii) clinical translation, as well as a third session of presentations from young investigators, which focused on recent achievements within Fondazione IRCCS INT on immunotherapy and targeted therapies. Presentations in the first session addressed the issue of cancer immunotherapy activity with respect to tumor heterogeneity, with key topics addressing: 1) tumor heterogeneity and targeted therapy, with the definition of the evolutionary Index as an indicator of tumor heterogeneity in both space and time; 2) the analysis of cancer evolution, with the introduction of the TRACERx Consortium-a multi-million pound UK research project focused on non-small cell lung cancer (NSCLC); 3) the use of anti-estrogen agents to boost immune recognition of breast cancer cells; and 4) the high degree of functional plasticity within the NK cell repertoire, including the expansion of adaptive NK cells following viral challenges. The second session addressed: 1) the effectiveness of radiotherapy to enhance the proportion of patients responsive to immune-checkpoint blockers (ICBs); 2) the use of MDSC scores in selecting melanoma patients with high probability to be responsive to ICBs; and 3) the relevance of the gut microbiome as a predictive factor, and the potential of its perturbation in increasing the immune response rate to ICBs. Overall, a picture emerged of tumor heterogeneity as the main limitation that impairs the effectiveness of anti-cancer therapies. Thus, the choice of a specific therapy based on reproducible and selective predictive biomarkers is an urgent unmet clinical need that should be addressed in order to increase the proportion of long-term responding patients and to improve the sustainability of novel drugs.
Collapse
Affiliation(s)
- F De Santis
- Immunotherapy and Innovative Therapeutics Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - M Del Vecchio
- Immunotherapy and Innovative Therapeutics Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Unit of Melanoma Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - L Castagnoli
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - F De Braud
- Medical Oncology Unit, Dept of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - S Di Cosimo
- Department of Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - D Franceschini
- Radiotherapy and Radiosurgery, Humanitas Clinical and Research Center, Via Manzoni 56 20089 Rozzano (Milano) Italy
| | - G Fucà
- Medical Oncology Unit, Dept of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - J Hiscott
- Laboratorio Pasteur, Istituto Pasteur-Fondazione Cenci-Bolognetti, 00161 Rome, Italy
| | - K J Malmberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden; Department. of Cellular Therapy and Allogeneic Stem Cell Transplantation, Karolinska University Hospital, Stockholm, Sweden; Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; The KG Jebsen Centre for Cancer Immunotherapy, University of Oslo, Oslo, Norway
| | - N McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - F Pietrantonio
- Medical Oncology Unit, Dept of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - L Rivoltini
- Unit of Immunotherapy of Human Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - S Sangaletti
- Molecular Immunology Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - E Tagliabue
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - C Tripodo
- Tumor Immunology Unit, Department of Health Science, Human Pathology Section, University of Palermo School of Medicine, Palermo, Italy
| | - C Vernieri
- Thoracic Oncology, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Fondazione Istituto FIRC di Oncologia Molecolare (IFOM), Milan, Italy
| | - L Zitvogel
- Gustave Roussy Cancer Campus (GRCC), Villejuif, France; Institut National de la Santé Et de la Recherche Medicale (INSERM), Villejuif, France; Univ. Paris-Sud, Université Paris-Saclay, Gustave Roussy, Villejuif, France; Center of Clinical Investigations in Biotherapies of Cancer (CICBT), Villejuif, France
| | - S M Pupa
- Molecular Targeting Unit, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - M Di Nicola
- Immunotherapy and Innovative Therapeutics Unit, Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy; Medical Oncology Unit, Dept of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| |
Collapse
|
2495
|
Weng YM, Peng M, Hu MX, Yao Y, Song QB. Clinical and molecular characteristics associated with the efficacy of PD-1/PD-L1 inhibitors for solid tumors: a meta-analysis. Onco Targets Ther 2018; 11:7529-7542. [PMID: 30464501 PMCID: PMC6214579 DOI: 10.2147/ott.s167865] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
We conducted a meta-analysis to estimate the impact of different clinical and molecular characteristics on the efficacy of programmed cell death 1 (PD-1) or programmed cell death ligand 1 (PD-L1) inhibitors. PubMed and Web of Science were searched for related trials. Eleven eligible studies, comprising 5,663 patients, were included in this meta-analysis. We found that the PD-1/PD-L1 inhibitor was associated with a 31% reduction in the risk of death (hazard ratio [HR]=0.69; 95% CI 0.64-0.74; P<0.00001) for patients with melanoma, non-small-cell lung cancer (NSCLC), urothelial carcinoma, head and neck carcinoma, and renal cell carcinoma. In subgroup analyses, all the patients with PD-L1-positive tumors had overall survival (OS) benefits from PD-1/PD-L1 inhibitors regardless of PD-L1 expression level, and a dose-effect relationship between the expression of PD-L1 and OS benefit from PD-1/PD-L1 inhibitors was observed. There was an OS improvement for patients with a smoking history (P<0.00001), but no OS benefit was observed for nonsmokers (P=0.28). In addition, first-line therapy had better OS than second-line or later treatment (P=0.02). No significant improvement of OS was observed (P=0.70) in patients aged ≥75 years. The relative treatment efficacy was similar according to sex (male vs female, P=0.60), performance status (0 vs ≥1, P=0.68), tumor histology (squamous NSCLC vs non-squamous NSCLC vs melanoma vs urothelial carcinoma vs head and neck carcinoma vs renal cell carcinoma, P=0.64), and treatment type (PD-1 inhibitor vs PD-L1 inhibitor, P=0.36). In conclusion, PD-L1-positive tumors, smoking history, and first-line treatment were potential factors for the efficacy of PD-1/PD-L1 inhibitors. Patients with higher PD-L1 expression might achieve greater OS benefits. In addition, sex, performance status, tumor histology, and treatment type could not predict the efficacy of this therapy. In contrast, patients aged >75 years and nonsmokers might not get OS benefits from this treatment. These results may improve treatment strategies and patient selection for PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Yi Ming Weng
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, People's Republic of China,
| | - Min Peng
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, People's Republic of China,
| | - Meng Xue Hu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, People's Republic of China,
| | - Yi Yao
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, People's Republic of China,
| | - Qi Bin Song
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, People's Republic of China,
| |
Collapse
|
2496
|
Davar D, Wang H, Chauvin JM, Pagliano O, Fourcade JJ, Ka M, Menna C, Rose A, Sander C, Borhani AA, Karunamurthy A, Tarhini AA, Tawbi HA, Zhao Q, Moreno BH, Ebbinghaus S, Ibrahim N, Kirkwood JM, Zarour HM. Phase Ib/II Study of Pembrolizumab and Pegylated-Interferon Alfa-2b in Advanced Melanoma. J Clin Oncol 2018; 36:JCO1800632. [PMID: 30359157 PMCID: PMC6286160 DOI: 10.1200/jco.18.00632] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
PURPOSE Objective responses are reported in 34% to 37% of patients with programmed death-1 (PD-1)-naïve advanced melanoma treated with PD-1 inhibitors. Pre-existing CD8+ T-cell infiltrate and interferon (IFN) gene signature correlate with response to PD-1 blockade. Here, we report a phase Ib/II study of pembrolizumab/pegylated (PEG)-IFN combination in PD-1-naïve advanced melanoma. PATIENTS AND METHODS PEG-IFN (1, 2, and 3 μg/kg per week) was dose escalated using a modified toxicity probability interval design in three cohorts of four patients each, whereas pembrolizumab was dosed at 2 mg/kg every 3 weeks in the phase Ib portion. Thirty-one patients were enrolled in the phase II portion. Primary objectives were safety and incidence of dose-limiting toxicities. Secondary objectives included objective response rate, progression-free survival (PFS), and overall survival. RESULTS Forty-three patients with stage IV melanoma were enrolled in the phase Ib and II portions of the study and included in the analysis. At the data cutoff date (December 31, 2017), median follow-up duration was 25 months (range, 1 to 38 months). All 43 patients experienced at least one adverse event; grade 3/4 treatment-related adverse events occurred in 21 of 43 patients (48.8%). Objective responses were seen at all three dose levels among 43 evaluable patients. The objective response rate was 60.5%, with 46.5% of patients exhibiting ongoing response. Median PFS was 11.0 months in all patients and unreached in responders, whereas median overall survival remained unreached in all patients. The 2-year PFS rate was 46%. CONCLUSION Pembrolizumab/PEG-IFN demonstrated an acceptable toxicity profile with promising evidence of clinical efficacy in PD-1-naïve metastatic melanoma. These results support the rationale to further investigate this pembrolizumab/PEG-IFN combination in this disease.
Collapse
Affiliation(s)
- Diwakar Davar
- Diwakar Davar, Hong Wang, Joe-Marc Chauvin, Ornella Pagliano, Julien J. Fourcade, Mignane Ka, Carmine Menna, Amy Rose, Cindy Sander, Amir A. Borhani, Arivarasan Karunamurthy, John M. Kirkwood, and Hassane M. Zarour, University of Pittsburgh, Pittsburgh, PA; Ahmad A. Tarhini, Cleveland Clinic, Cleveland, OH; Hussein A. Tawbi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Qing Zhao, Blanca H. Moreno, Scott Ebbinghaus, and Nageatte Ibrahim, Merck, Kenilworth, NJ
| | - Hong Wang
- Diwakar Davar, Hong Wang, Joe-Marc Chauvin, Ornella Pagliano, Julien J. Fourcade, Mignane Ka, Carmine Menna, Amy Rose, Cindy Sander, Amir A. Borhani, Arivarasan Karunamurthy, John M. Kirkwood, and Hassane M. Zarour, University of Pittsburgh, Pittsburgh, PA; Ahmad A. Tarhini, Cleveland Clinic, Cleveland, OH; Hussein A. Tawbi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Qing Zhao, Blanca H. Moreno, Scott Ebbinghaus, and Nageatte Ibrahim, Merck, Kenilworth, NJ
| | - Joe-Marc Chauvin
- Diwakar Davar, Hong Wang, Joe-Marc Chauvin, Ornella Pagliano, Julien J. Fourcade, Mignane Ka, Carmine Menna, Amy Rose, Cindy Sander, Amir A. Borhani, Arivarasan Karunamurthy, John M. Kirkwood, and Hassane M. Zarour, University of Pittsburgh, Pittsburgh, PA; Ahmad A. Tarhini, Cleveland Clinic, Cleveland, OH; Hussein A. Tawbi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Qing Zhao, Blanca H. Moreno, Scott Ebbinghaus, and Nageatte Ibrahim, Merck, Kenilworth, NJ
| | - Ornella Pagliano
- Diwakar Davar, Hong Wang, Joe-Marc Chauvin, Ornella Pagliano, Julien J. Fourcade, Mignane Ka, Carmine Menna, Amy Rose, Cindy Sander, Amir A. Borhani, Arivarasan Karunamurthy, John M. Kirkwood, and Hassane M. Zarour, University of Pittsburgh, Pittsburgh, PA; Ahmad A. Tarhini, Cleveland Clinic, Cleveland, OH; Hussein A. Tawbi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Qing Zhao, Blanca H. Moreno, Scott Ebbinghaus, and Nageatte Ibrahim, Merck, Kenilworth, NJ
| | - Julien J. Fourcade
- Diwakar Davar, Hong Wang, Joe-Marc Chauvin, Ornella Pagliano, Julien J. Fourcade, Mignane Ka, Carmine Menna, Amy Rose, Cindy Sander, Amir A. Borhani, Arivarasan Karunamurthy, John M. Kirkwood, and Hassane M. Zarour, University of Pittsburgh, Pittsburgh, PA; Ahmad A. Tarhini, Cleveland Clinic, Cleveland, OH; Hussein A. Tawbi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Qing Zhao, Blanca H. Moreno, Scott Ebbinghaus, and Nageatte Ibrahim, Merck, Kenilworth, NJ
| | - Mignane Ka
- Diwakar Davar, Hong Wang, Joe-Marc Chauvin, Ornella Pagliano, Julien J. Fourcade, Mignane Ka, Carmine Menna, Amy Rose, Cindy Sander, Amir A. Borhani, Arivarasan Karunamurthy, John M. Kirkwood, and Hassane M. Zarour, University of Pittsburgh, Pittsburgh, PA; Ahmad A. Tarhini, Cleveland Clinic, Cleveland, OH; Hussein A. Tawbi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Qing Zhao, Blanca H. Moreno, Scott Ebbinghaus, and Nageatte Ibrahim, Merck, Kenilworth, NJ
| | - Carmine Menna
- Diwakar Davar, Hong Wang, Joe-Marc Chauvin, Ornella Pagliano, Julien J. Fourcade, Mignane Ka, Carmine Menna, Amy Rose, Cindy Sander, Amir A. Borhani, Arivarasan Karunamurthy, John M. Kirkwood, and Hassane M. Zarour, University of Pittsburgh, Pittsburgh, PA; Ahmad A. Tarhini, Cleveland Clinic, Cleveland, OH; Hussein A. Tawbi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Qing Zhao, Blanca H. Moreno, Scott Ebbinghaus, and Nageatte Ibrahim, Merck, Kenilworth, NJ
| | - Amy Rose
- Diwakar Davar, Hong Wang, Joe-Marc Chauvin, Ornella Pagliano, Julien J. Fourcade, Mignane Ka, Carmine Menna, Amy Rose, Cindy Sander, Amir A. Borhani, Arivarasan Karunamurthy, John M. Kirkwood, and Hassane M. Zarour, University of Pittsburgh, Pittsburgh, PA; Ahmad A. Tarhini, Cleveland Clinic, Cleveland, OH; Hussein A. Tawbi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Qing Zhao, Blanca H. Moreno, Scott Ebbinghaus, and Nageatte Ibrahim, Merck, Kenilworth, NJ
| | - Cindy Sander
- Diwakar Davar, Hong Wang, Joe-Marc Chauvin, Ornella Pagliano, Julien J. Fourcade, Mignane Ka, Carmine Menna, Amy Rose, Cindy Sander, Amir A. Borhani, Arivarasan Karunamurthy, John M. Kirkwood, and Hassane M. Zarour, University of Pittsburgh, Pittsburgh, PA; Ahmad A. Tarhini, Cleveland Clinic, Cleveland, OH; Hussein A. Tawbi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Qing Zhao, Blanca H. Moreno, Scott Ebbinghaus, and Nageatte Ibrahim, Merck, Kenilworth, NJ
| | - Amir A. Borhani
- Diwakar Davar, Hong Wang, Joe-Marc Chauvin, Ornella Pagliano, Julien J. Fourcade, Mignane Ka, Carmine Menna, Amy Rose, Cindy Sander, Amir A. Borhani, Arivarasan Karunamurthy, John M. Kirkwood, and Hassane M. Zarour, University of Pittsburgh, Pittsburgh, PA; Ahmad A. Tarhini, Cleveland Clinic, Cleveland, OH; Hussein A. Tawbi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Qing Zhao, Blanca H. Moreno, Scott Ebbinghaus, and Nageatte Ibrahim, Merck, Kenilworth, NJ
| | - Arivarasan Karunamurthy
- Diwakar Davar, Hong Wang, Joe-Marc Chauvin, Ornella Pagliano, Julien J. Fourcade, Mignane Ka, Carmine Menna, Amy Rose, Cindy Sander, Amir A. Borhani, Arivarasan Karunamurthy, John M. Kirkwood, and Hassane M. Zarour, University of Pittsburgh, Pittsburgh, PA; Ahmad A. Tarhini, Cleveland Clinic, Cleveland, OH; Hussein A. Tawbi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Qing Zhao, Blanca H. Moreno, Scott Ebbinghaus, and Nageatte Ibrahim, Merck, Kenilworth, NJ
| | - Ahmad A. Tarhini
- Diwakar Davar, Hong Wang, Joe-Marc Chauvin, Ornella Pagliano, Julien J. Fourcade, Mignane Ka, Carmine Menna, Amy Rose, Cindy Sander, Amir A. Borhani, Arivarasan Karunamurthy, John M. Kirkwood, and Hassane M. Zarour, University of Pittsburgh, Pittsburgh, PA; Ahmad A. Tarhini, Cleveland Clinic, Cleveland, OH; Hussein A. Tawbi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Qing Zhao, Blanca H. Moreno, Scott Ebbinghaus, and Nageatte Ibrahim, Merck, Kenilworth, NJ
| | - Hussein A. Tawbi
- Diwakar Davar, Hong Wang, Joe-Marc Chauvin, Ornella Pagliano, Julien J. Fourcade, Mignane Ka, Carmine Menna, Amy Rose, Cindy Sander, Amir A. Borhani, Arivarasan Karunamurthy, John M. Kirkwood, and Hassane M. Zarour, University of Pittsburgh, Pittsburgh, PA; Ahmad A. Tarhini, Cleveland Clinic, Cleveland, OH; Hussein A. Tawbi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Qing Zhao, Blanca H. Moreno, Scott Ebbinghaus, and Nageatte Ibrahim, Merck, Kenilworth, NJ
| | - Qing Zhao
- Diwakar Davar, Hong Wang, Joe-Marc Chauvin, Ornella Pagliano, Julien J. Fourcade, Mignane Ka, Carmine Menna, Amy Rose, Cindy Sander, Amir A. Borhani, Arivarasan Karunamurthy, John M. Kirkwood, and Hassane M. Zarour, University of Pittsburgh, Pittsburgh, PA; Ahmad A. Tarhini, Cleveland Clinic, Cleveland, OH; Hussein A. Tawbi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Qing Zhao, Blanca H. Moreno, Scott Ebbinghaus, and Nageatte Ibrahim, Merck, Kenilworth, NJ
| | - Blanca H. Moreno
- Diwakar Davar, Hong Wang, Joe-Marc Chauvin, Ornella Pagliano, Julien J. Fourcade, Mignane Ka, Carmine Menna, Amy Rose, Cindy Sander, Amir A. Borhani, Arivarasan Karunamurthy, John M. Kirkwood, and Hassane M. Zarour, University of Pittsburgh, Pittsburgh, PA; Ahmad A. Tarhini, Cleveland Clinic, Cleveland, OH; Hussein A. Tawbi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Qing Zhao, Blanca H. Moreno, Scott Ebbinghaus, and Nageatte Ibrahim, Merck, Kenilworth, NJ
| | - Scott Ebbinghaus
- Diwakar Davar, Hong Wang, Joe-Marc Chauvin, Ornella Pagliano, Julien J. Fourcade, Mignane Ka, Carmine Menna, Amy Rose, Cindy Sander, Amir A. Borhani, Arivarasan Karunamurthy, John M. Kirkwood, and Hassane M. Zarour, University of Pittsburgh, Pittsburgh, PA; Ahmad A. Tarhini, Cleveland Clinic, Cleveland, OH; Hussein A. Tawbi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Qing Zhao, Blanca H. Moreno, Scott Ebbinghaus, and Nageatte Ibrahim, Merck, Kenilworth, NJ
| | - Nageatte Ibrahim
- Diwakar Davar, Hong Wang, Joe-Marc Chauvin, Ornella Pagliano, Julien J. Fourcade, Mignane Ka, Carmine Menna, Amy Rose, Cindy Sander, Amir A. Borhani, Arivarasan Karunamurthy, John M. Kirkwood, and Hassane M. Zarour, University of Pittsburgh, Pittsburgh, PA; Ahmad A. Tarhini, Cleveland Clinic, Cleveland, OH; Hussein A. Tawbi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Qing Zhao, Blanca H. Moreno, Scott Ebbinghaus, and Nageatte Ibrahim, Merck, Kenilworth, NJ
| | - John M. Kirkwood
- Diwakar Davar, Hong Wang, Joe-Marc Chauvin, Ornella Pagliano, Julien J. Fourcade, Mignane Ka, Carmine Menna, Amy Rose, Cindy Sander, Amir A. Borhani, Arivarasan Karunamurthy, John M. Kirkwood, and Hassane M. Zarour, University of Pittsburgh, Pittsburgh, PA; Ahmad A. Tarhini, Cleveland Clinic, Cleveland, OH; Hussein A. Tawbi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Qing Zhao, Blanca H. Moreno, Scott Ebbinghaus, and Nageatte Ibrahim, Merck, Kenilworth, NJ
| | - Hassane M. Zarour
- Diwakar Davar, Hong Wang, Joe-Marc Chauvin, Ornella Pagliano, Julien J. Fourcade, Mignane Ka, Carmine Menna, Amy Rose, Cindy Sander, Amir A. Borhani, Arivarasan Karunamurthy, John M. Kirkwood, and Hassane M. Zarour, University of Pittsburgh, Pittsburgh, PA; Ahmad A. Tarhini, Cleveland Clinic, Cleveland, OH; Hussein A. Tawbi, The University of Texas MD Anderson Cancer Center, Houston, TX; and Qing Zhao, Blanca H. Moreno, Scott Ebbinghaus, and Nageatte Ibrahim, Merck, Kenilworth, NJ
| |
Collapse
|
2497
|
Affiliation(s)
- Mark J Smyth
- Immunology in Cancer and Infection Laboratory QIMR Berghofer Medical Research Institute 300 Herston Road Herston 4006 QLD Australia
| | - Michele Wl Teng
- Cancer Immunoregulation and Immunotherapy QIMR Berghofer Medical Research Institute 300 Herston Road Herston 4006 QLD Australia
| |
Collapse
|
2498
|
Elias R, Hartshorn K, Rahma O, Lin N, Snyder-Cappione JE. Aging, immune senescence, and immunotherapy: A comprehensive review. Semin Oncol 2018; 45:187-200. [PMID: 30539714 DOI: 10.1053/j.seminoncol.2018.08.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 07/07/2018] [Accepted: 08/01/2018] [Indexed: 12/22/2022]
Abstract
The advent of immune checkpoint inhibitors (ICIs) has changed the landscape of cancer treatment. Older adults represent the majority of cancer patients; however, direct data evaluating ICIs in this patient population is lacking. Aging is associated with changes in the immune system known as "immunosenescence" that could impact the efficacy and safety profile of ICIs. In this paper, we review aging-associated changes in the immune system as they may relate to cancer and immunotherapy, with mention of the effect of chronic viral infections and frailty. Furthermore, we summarize the current clinical evidence of ICI effectiveness and toxicity among older adults with cancer.
Collapse
Affiliation(s)
- Rawad Elias
- Hartford HealthCare Cancer Institute, Hartford Hospital, Hartford, CT, USA.
| | - Kevan Hartshorn
- Section of Hematology Oncology, Boston University School of Medicine, Boston, MA, USA
| | - Osama Rahma
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Nina Lin
- Department of Medicine, Boston Medical Center, Boston University School of Medicine, MA, USA
| | - Jennifer E Snyder-Cappione
- Department of Microbiology, Boston University School of Medicine, Boston, MA, USA; Flow Cytometry Core Facility, Boston University School of Medicine, Boston, MA, USA
| |
Collapse
|
2499
|
Lu JW, Miao Y, Guo CX, Ke QF, Yin JH, Zhou SM, Guo YP. Lanthanum-Doped Chitosan Hydrogels Promote the Apoptosis of Melanoma Cells by Bcl-2/Bax Pathway. ACS APPLIED BIO MATERIALS 2018; 1:1468-1477. [DOI: 10.1021/acsabm.8b00417] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Jia-Wei Lu
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, China
| | | | - Cui-Xiang Guo
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, China
| | - Qin-Fei Ke
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, China
| | | | | | - Ya-Ping Guo
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, China
| |
Collapse
|
2500
|
Tarhini A, Lin Y, Lin H, Rahman Z, Vallabhaneni P, Mendiratta P, Pingpank JF, Holtzman MP, Yusko EC, Rytlewski JA, Rao UNM, Ferris RL, Kirkwood JM. Neoadjuvant ipilimumab (3 mg/kg or 10 mg/kg) and high dose IFN-α2b in locally/regionally advanced melanoma: safety, efficacy and impact on T-cell repertoire. J Immunother Cancer 2018; 6:112. [PMID: 30352626 PMCID: PMC6199801 DOI: 10.1186/s40425-018-0428-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 10/10/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Neoadjuvant immunotherapy utilizing novel combinations has the potential to transform the standard of care for locally/regionally advanced melanoma. We hypothesized that neoadjuvant ipilimumab in combination with high dose IFNα2b (HDI) is safe and associated with durable pathologic complete responses (pCR). METHODS Patients with locally/regionally advanced melanoma were randomized to ipilimumab 3 or 10 mg/kg × 4 doses bracketing definitive surgery, then every 12 weeks × 4. HDI was given concurrently. We evaluated the safety and efficacy of the combination with ipilimumab 3 or 10 mg/kg. The impact on T-cell fraction and clonality were investigated in tumor and blood. RESULTS Thirty patients (age 37-76), 15 each at 3 and 10 mg/kg, 18 male and 12 female were treated. Considering immune related adverse events (irAEs) of interest, more grade 3/4 irAEs were seen with ipilimumab 10 mg/kg versus 3 mg/kg (p = 0.042). Among 28 evaluable patients, 11 relapsed, of whom 5 died. Median follow-up for 17 patients who have not relapsed was 32 months. The radiologic preoperative response rate was 36% (95% CI, 21-54); 4 patients at ipilimumab 3 mg/kg and 6 at 10 mg/kg and 2 (at 10 mg/kg) later relapsed. The pCR was 32% (95% CI, 18-51); 5 patients at ipilimumab 3 mg/kg and 4 at 10 mg/kg and one (at 3 mg/kg) had a late relapse. In patients with pCR, T-cell fraction was significantly higher when measured in primary melanoma tumors (p = 0.033). Higher tumor T-cell clonality in primary tumor and more so following neoadjuvant therapy was significantly associated with improved relapse free survival. CONCLUSIONS Neoadjuvant ipilimumab-HDI was relatively safe and exhibited promising tumor response rates with an associated measurable impact on T-cell fraction and clonality. Most pCRs were durable supporting the value of pCR as a primary endpoint in neoadjuvant immunotherapy trials. TRIAL REGISTRATION ClinicalTrials.gov, NCT01608594 . Registered 31 May 2012.
Collapse
Affiliation(s)
- Ahmad Tarhini
- UPMC Hillman Cancer Center, Pittsburgh, USA. .,Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Institute and Case Comprehensive Cancer Center, 9500 Euclid Ave CA6-157, Cleveland, OH, 44195, USA.
| | - Yan Lin
- UPMC Hillman Cancer Center, Pittsburgh, USA
| | - Huang Lin
- UPMC Hillman Cancer Center, Pittsburgh, USA
| | | | | | - Prateek Mendiratta
- Department of Hematology and Oncology, Cleveland Clinic Taussig Cancer Institute and Case Comprehensive Cancer Center, 9500 Euclid Ave CA6-157, Cleveland, OH, 44195, USA
| | | | | | | | | | | | | | | |
Collapse
|