151
|
Segal NH, Ou SHI, Balmanoukian A, Fury MG, Massarelli E, Brahmer JR, Weiss J, Schöffski P, Antonia SJ, Massard C, Zandberg DP, Khleif SN, Xiao F, Rebelatto MC, Steele KE, Robbins PB, Angra N, Song X, Abdullah S, Butler M. Safety and efficacy of durvalumab in patients with head and neck squamous cell carcinoma: results from a phase I/II expansion cohort. Eur J Cancer 2019; 109:154-161. [PMID: 30731276 DOI: 10.1016/j.ejca.2018.12.029] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 11/30/2018] [Accepted: 12/26/2018] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Durvalumab selectively blocks programmed cell death ligand-1 (PD-L1) binding to programmed cell death-1. Encouraging clinical activity and manageable safety were reported in urothelial carcinoma, non-small-cell lung cancer (NSCLC), hepatocellular carcinoma (HC) and small-cell lung cancer (SCLC) in a multicenter phase I/II study. Safety and clinical activity in recurrent/metastatic head and neck squamous cell carcinoma (HNSCC) were evaluated in the expansion phase. METHODS Patients received 10 mg/kg of durvalumab intravenously every 2 weeks for 12 months or until confirmed progressive disease or unacceptable toxicity. The primary objective was safety; clinical activity was a secondary objective. RESULTS Sixty-two patients were enrolled and evaluable (received first dose ≥24 weeks before data cutoff). Median age was 57 years; 40.3% were human papillomavirus (HPV)-positive; 32.3% had tumour cell PD-L1 expression ≥25%, and 62.9% were current/former smokers. They had a median of 2 prior systemic treatments (range, 1-13). All-causality adverse events (AEs) occurred in 98.4%; drug-related AEs occurred in 59.7% and were grade III-IV in 9.7%. There were no drug-related discontinuations or deaths. Objective response rate (blinded independent central review) was 6.5% (15.0% for PD-L1 ≥25%, 2.6% for <25%). Median time to response was 2.7 months (range, 1.2-5.5); median duration was 12.4 months (range, 3.5-20.5+). Median progression-free survival was 1.4 months; median overall survival (OS) was 8.4 months. OS rate was 62% at 6 months and 38% at 12 months (42% for PD-L1 ≥25%, 36% for <25%). CONCLUSIONS Durvalumab safety in HNSCC was manageable and consistent with other cohorts of the study. Early, durable responses in these heavily pretreated patients warrant further investigation; phase III monotherapy and combination therapy studies are ongoing. CLINICAL TRIAL REGISTRY: clinicaltrials.gov NCT01693562; MedImmune study 1108.
Collapse
Affiliation(s)
- Neil H Segal
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| | - Sai-Hong I Ou
- Chao Family Comprehensive Cancer Center, Department of Medicine, Division of Hematology/Oncology, University of California School of Medicine, Orange, CA, USA
| | - Ani Balmanoukian
- Hematology/Oncology, The Angeles Clinic and Research Institute, Los Angeles, CA, USA
| | - Matthew G Fury
- Department of Medicine, Head and Neck Medical Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Erminia Massarelli
- Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Julie R Brahmer
- Thoracic Oncology Program, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Jared Weiss
- Division of Hematology/Oncology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Patrick Schöffski
- Department of Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Scott J Antonia
- Department of Thoracic Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Christophe Massard
- Université Paris Saclay, Université Paris-Sud, Drug Development Department, Gustave Roussy, Villejuif, France
| | - Dan P Zandberg
- Head and Neck and Thyroid Cancer Disease Sections, University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
| | - Samir N Khleif
- Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Feng Xiao
- Biostatistics, MedImmune, Gaithersburg, MD, USA
| | | | - Keith E Steele
- Translational Sciences, MedImmune, Gaithersburg, MD, USA
| | - Paul B Robbins
- Translational Sciences, MedImmune, Gaithersburg, MD, USA
| | - Natasha Angra
- Clinical Development, MedImmune, Gaithersburg, MD, USA
| | - Xuyang Song
- Translational Sciences, MedImmune, Gaithersburg, MD, USA
| | | | - Marcus Butler
- Tumor Immunotherapy Program, Princess Margaret Cancer Centre, Toronto, ON, Canada
| |
Collapse
|
152
|
Laurie SA, Banerji S, Blais N, Brule S, Cheema PK, Cheung P, Daaboul N, Hao D, Hirsh V, Juergens R, Laskin J, Leighl N, MacRae R, Nicholas G, Roberge D, Rothenstein J, Stewart DJ, Tsao MS. Canadian consensus: oligoprogressive, pseudoprogressive, and oligometastatic non-small-cell lung cancer. Curr Oncol 2019; 26:e81-e93. [PMID: 30853813 PMCID: PMC6380642 DOI: 10.3747/co.26.4116] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background Little evidence has been generated for how best to manage patients with non-small-cell lung cancer (nsclc) presenting with rarer clinical scenarios, including oligometastases, oligoprogression, and pseudoprogression. In each of those scenarios, oncologists have to consider how best to balance efficacy with quality of life, while maximizing the duration of each line of therapy and ensuring that patients are still eligible for later options, including clinical trial enrolment. Methods An expert panel was convened to define the clinical questions. Using case-based presentations, consensus practice recommendations for each clinical scenario were generated through focused, evidence-based discussions. Results Treatment strategies and best-practice or consensus recommendations are presented, with areas of consensus and areas of uncertainty identified. Conclusions In each situation, treatment has to be tailored to suit the individual patient, but with the intent of extending and maximizing the use of each line of treatment, while keeping treatment options in reserve for later lines of therapy. Patient participation in clinical trials examining these issues should be encouraged.
Collapse
Affiliation(s)
- S A Laurie
- Ontario: The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa (Laurie); Division of Medical Oncology, The Ottawa Hospital, University of Ottawa, Ottawa (Brule); University of Toronto, Toronto, and William Osler Health System, Brampton (Cheema); Sunnybrook Odette Cancer Centre, Department of Radiation Oncology, University of Toronto, Toronto (Cheung); McMaster University, Juravinski Cancer Centre, Hamilton (Juergens); Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto (Leighl); University of Ottawa, The Ottawa Hospital, Ottawa (MacRae); University of Ottawa, Ottawa (Nicholas); R.S. McLaughlin Durham Regional Cancer Centre, Lakeridge Health, Oshawa, and Queen's University, Kingston (Rothenstein); The Ottawa Hospital, The Ottawa Hospital Research Institute, and Division of Medical Oncology, University of Ottawa, Ottawa (Stewart); University Health Network, Princess Margaret Cancer Centre, and University of Toronto, Toronto (Tsao)
| | - S Banerji
- Manitoba: Rady Faculty of Health Sciences, University of Manitoba, and Medical Oncology, CancerCare Manitoba, Winnipeg
| | - N Blais
- Quebec: CHUM Cancer Centre, Université de Montréal, Montreal (Blais); Centre intégré de cancérologie de la Montérégie, Hôpital Charles-LeMoyne, and Université de Sherbrooke, Greenfield Park (Daaboul); Department of Oncology, McGill University, and Thoracic Oncology, McGill University Health Centre, Montreal (Hirsh); Centre hospitalier de l'Université de Montréal, Montreal (Roberge)
| | - S Brule
- Ontario: The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa (Laurie); Division of Medical Oncology, The Ottawa Hospital, University of Ottawa, Ottawa (Brule); University of Toronto, Toronto, and William Osler Health System, Brampton (Cheema); Sunnybrook Odette Cancer Centre, Department of Radiation Oncology, University of Toronto, Toronto (Cheung); McMaster University, Juravinski Cancer Centre, Hamilton (Juergens); Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto (Leighl); University of Ottawa, The Ottawa Hospital, Ottawa (MacRae); University of Ottawa, Ottawa (Nicholas); R.S. McLaughlin Durham Regional Cancer Centre, Lakeridge Health, Oshawa, and Queen's University, Kingston (Rothenstein); The Ottawa Hospital, The Ottawa Hospital Research Institute, and Division of Medical Oncology, University of Ottawa, Ottawa (Stewart); University Health Network, Princess Margaret Cancer Centre, and University of Toronto, Toronto (Tsao)
| | - P K Cheema
- Ontario: The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa (Laurie); Division of Medical Oncology, The Ottawa Hospital, University of Ottawa, Ottawa (Brule); University of Toronto, Toronto, and William Osler Health System, Brampton (Cheema); Sunnybrook Odette Cancer Centre, Department of Radiation Oncology, University of Toronto, Toronto (Cheung); McMaster University, Juravinski Cancer Centre, Hamilton (Juergens); Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto (Leighl); University of Ottawa, The Ottawa Hospital, Ottawa (MacRae); University of Ottawa, Ottawa (Nicholas); R.S. McLaughlin Durham Regional Cancer Centre, Lakeridge Health, Oshawa, and Queen's University, Kingston (Rothenstein); The Ottawa Hospital, The Ottawa Hospital Research Institute, and Division of Medical Oncology, University of Ottawa, Ottawa (Stewart); University Health Network, Princess Margaret Cancer Centre, and University of Toronto, Toronto (Tsao)
| | - P Cheung
- Ontario: The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa (Laurie); Division of Medical Oncology, The Ottawa Hospital, University of Ottawa, Ottawa (Brule); University of Toronto, Toronto, and William Osler Health System, Brampton (Cheema); Sunnybrook Odette Cancer Centre, Department of Radiation Oncology, University of Toronto, Toronto (Cheung); McMaster University, Juravinski Cancer Centre, Hamilton (Juergens); Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto (Leighl); University of Ottawa, The Ottawa Hospital, Ottawa (MacRae); University of Ottawa, Ottawa (Nicholas); R.S. McLaughlin Durham Regional Cancer Centre, Lakeridge Health, Oshawa, and Queen's University, Kingston (Rothenstein); The Ottawa Hospital, The Ottawa Hospital Research Institute, and Division of Medical Oncology, University of Ottawa, Ottawa (Stewart); University Health Network, Princess Margaret Cancer Centre, and University of Toronto, Toronto (Tsao)
| | - N Daaboul
- Quebec: CHUM Cancer Centre, Université de Montréal, Montreal (Blais); Centre intégré de cancérologie de la Montérégie, Hôpital Charles-LeMoyne, and Université de Sherbrooke, Greenfield Park (Daaboul); Department of Oncology, McGill University, and Thoracic Oncology, McGill University Health Centre, Montreal (Hirsh); Centre hospitalier de l'Université de Montréal, Montreal (Roberge)
| | - D Hao
- Alberta: Tom Baker Cancer Centre and Department of Oncology, University of Calgary, Calgary
| | - V Hirsh
- Quebec: CHUM Cancer Centre, Université de Montréal, Montreal (Blais); Centre intégré de cancérologie de la Montérégie, Hôpital Charles-LeMoyne, and Université de Sherbrooke, Greenfield Park (Daaboul); Department of Oncology, McGill University, and Thoracic Oncology, McGill University Health Centre, Montreal (Hirsh); Centre hospitalier de l'Université de Montréal, Montreal (Roberge)
| | - R Juergens
- Ontario: The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa (Laurie); Division of Medical Oncology, The Ottawa Hospital, University of Ottawa, Ottawa (Brule); University of Toronto, Toronto, and William Osler Health System, Brampton (Cheema); Sunnybrook Odette Cancer Centre, Department of Radiation Oncology, University of Toronto, Toronto (Cheung); McMaster University, Juravinski Cancer Centre, Hamilton (Juergens); Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto (Leighl); University of Ottawa, The Ottawa Hospital, Ottawa (MacRae); University of Ottawa, Ottawa (Nicholas); R.S. McLaughlin Durham Regional Cancer Centre, Lakeridge Health, Oshawa, and Queen's University, Kingston (Rothenstein); The Ottawa Hospital, The Ottawa Hospital Research Institute, and Division of Medical Oncology, University of Ottawa, Ottawa (Stewart); University Health Network, Princess Margaret Cancer Centre, and University of Toronto, Toronto (Tsao)
| | - J Laskin
- British Columbia: Medical Oncology, BC Cancer, Vancouver
| | - N Leighl
- Ontario: The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa (Laurie); Division of Medical Oncology, The Ottawa Hospital, University of Ottawa, Ottawa (Brule); University of Toronto, Toronto, and William Osler Health System, Brampton (Cheema); Sunnybrook Odette Cancer Centre, Department of Radiation Oncology, University of Toronto, Toronto (Cheung); McMaster University, Juravinski Cancer Centre, Hamilton (Juergens); Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto (Leighl); University of Ottawa, The Ottawa Hospital, Ottawa (MacRae); University of Ottawa, Ottawa (Nicholas); R.S. McLaughlin Durham Regional Cancer Centre, Lakeridge Health, Oshawa, and Queen's University, Kingston (Rothenstein); The Ottawa Hospital, The Ottawa Hospital Research Institute, and Division of Medical Oncology, University of Ottawa, Ottawa (Stewart); University Health Network, Princess Margaret Cancer Centre, and University of Toronto, Toronto (Tsao)
| | - R MacRae
- Ontario: The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa (Laurie); Division of Medical Oncology, The Ottawa Hospital, University of Ottawa, Ottawa (Brule); University of Toronto, Toronto, and William Osler Health System, Brampton (Cheema); Sunnybrook Odette Cancer Centre, Department of Radiation Oncology, University of Toronto, Toronto (Cheung); McMaster University, Juravinski Cancer Centre, Hamilton (Juergens); Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto (Leighl); University of Ottawa, The Ottawa Hospital, Ottawa (MacRae); University of Ottawa, Ottawa (Nicholas); R.S. McLaughlin Durham Regional Cancer Centre, Lakeridge Health, Oshawa, and Queen's University, Kingston (Rothenstein); The Ottawa Hospital, The Ottawa Hospital Research Institute, and Division of Medical Oncology, University of Ottawa, Ottawa (Stewart); University Health Network, Princess Margaret Cancer Centre, and University of Toronto, Toronto (Tsao)
| | - G Nicholas
- Ontario: The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa (Laurie); Division of Medical Oncology, The Ottawa Hospital, University of Ottawa, Ottawa (Brule); University of Toronto, Toronto, and William Osler Health System, Brampton (Cheema); Sunnybrook Odette Cancer Centre, Department of Radiation Oncology, University of Toronto, Toronto (Cheung); McMaster University, Juravinski Cancer Centre, Hamilton (Juergens); Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto (Leighl); University of Ottawa, The Ottawa Hospital, Ottawa (MacRae); University of Ottawa, Ottawa (Nicholas); R.S. McLaughlin Durham Regional Cancer Centre, Lakeridge Health, Oshawa, and Queen's University, Kingston (Rothenstein); The Ottawa Hospital, The Ottawa Hospital Research Institute, and Division of Medical Oncology, University of Ottawa, Ottawa (Stewart); University Health Network, Princess Margaret Cancer Centre, and University of Toronto, Toronto (Tsao)
| | - D Roberge
- Quebec: CHUM Cancer Centre, Université de Montréal, Montreal (Blais); Centre intégré de cancérologie de la Montérégie, Hôpital Charles-LeMoyne, and Université de Sherbrooke, Greenfield Park (Daaboul); Department of Oncology, McGill University, and Thoracic Oncology, McGill University Health Centre, Montreal (Hirsh); Centre hospitalier de l'Université de Montréal, Montreal (Roberge)
| | - J Rothenstein
- Ontario: The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa (Laurie); Division of Medical Oncology, The Ottawa Hospital, University of Ottawa, Ottawa (Brule); University of Toronto, Toronto, and William Osler Health System, Brampton (Cheema); Sunnybrook Odette Cancer Centre, Department of Radiation Oncology, University of Toronto, Toronto (Cheung); McMaster University, Juravinski Cancer Centre, Hamilton (Juergens); Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto (Leighl); University of Ottawa, The Ottawa Hospital, Ottawa (MacRae); University of Ottawa, Ottawa (Nicholas); R.S. McLaughlin Durham Regional Cancer Centre, Lakeridge Health, Oshawa, and Queen's University, Kingston (Rothenstein); The Ottawa Hospital, The Ottawa Hospital Research Institute, and Division of Medical Oncology, University of Ottawa, Ottawa (Stewart); University Health Network, Princess Margaret Cancer Centre, and University of Toronto, Toronto (Tsao)
| | - D J Stewart
- Ontario: The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa (Laurie); Division of Medical Oncology, The Ottawa Hospital, University of Ottawa, Ottawa (Brule); University of Toronto, Toronto, and William Osler Health System, Brampton (Cheema); Sunnybrook Odette Cancer Centre, Department of Radiation Oncology, University of Toronto, Toronto (Cheung); McMaster University, Juravinski Cancer Centre, Hamilton (Juergens); Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto (Leighl); University of Ottawa, The Ottawa Hospital, Ottawa (MacRae); University of Ottawa, Ottawa (Nicholas); R.S. McLaughlin Durham Regional Cancer Centre, Lakeridge Health, Oshawa, and Queen's University, Kingston (Rothenstein); The Ottawa Hospital, The Ottawa Hospital Research Institute, and Division of Medical Oncology, University of Ottawa, Ottawa (Stewart); University Health Network, Princess Margaret Cancer Centre, and University of Toronto, Toronto (Tsao)
| | - M S Tsao
- Ontario: The Ottawa Hospital Cancer Centre, University of Ottawa, Ottawa (Laurie); Division of Medical Oncology, The Ottawa Hospital, University of Ottawa, Ottawa (Brule); University of Toronto, Toronto, and William Osler Health System, Brampton (Cheema); Sunnybrook Odette Cancer Centre, Department of Radiation Oncology, University of Toronto, Toronto (Cheung); McMaster University, Juravinski Cancer Centre, Hamilton (Juergens); Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto (Leighl); University of Ottawa, The Ottawa Hospital, Ottawa (MacRae); University of Ottawa, Ottawa (Nicholas); R.S. McLaughlin Durham Regional Cancer Centre, Lakeridge Health, Oshawa, and Queen's University, Kingston (Rothenstein); The Ottawa Hospital, The Ottawa Hospital Research Institute, and Division of Medical Oncology, University of Ottawa, Ottawa (Stewart); University Health Network, Princess Margaret Cancer Centre, and University of Toronto, Toronto (Tsao)
| |
Collapse
|
153
|
Enfield KSS, Martin SD, Marshall EA, Kung SHY, Gallagher P, Milne K, Chen Z, Nelson BH, Lam S, English JC, MacAulay CE, Lam WL, Guillaud M. Hyperspectral cell sociology reveals spatial tumor-immune cell interactions associated with lung cancer recurrence. J Immunother Cancer 2019; 7:13. [PMID: 30651131 PMCID: PMC6335759 DOI: 10.1186/s40425-018-0488-6] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 12/19/2018] [Indexed: 01/06/2023] Open
Abstract
Background The tumor microenvironment (TME) is a complex mixture of tumor epithelium, stroma and immune cells, and the immune component of the TME is highly prognostic for tumor progression and patient outcome. In lung cancer, anti-PD-1 therapy significantly improves patient survival through activation of T cell cytotoxicity against tumor cells. Direct contact between CD8+ T cells and target cells is necessary for CD8+ T cell activity, indicating that spatial organization of immune cells within the TME reflects a critical process in anti-tumor immunity. Current immunohistochemistry (IHC) imaging techniques identify immune cell numbers and densities, but lack assessment of cell–cell spatial relationships (or “cell sociology”). Immune functionality, however, is often dictated by cell-to-cell contact and cannot be resolved by simple metrics of cell density (for example, number of cells per mm2). To address this issue, we developed a Hyperspectral Cell Sociology technology platform for the analysis of cell–cell interactions in multi-channel IHC-stained tissue. Methods Tissue sections of primary tumors from lung adenocarcinoma patients with known clinical outcome were stained using multiplex IHC for CD3, CD8, and CD79a, and hyperspectral image analysis determined the phenotype of all cells. A Voronoi diagram for each cell was used to approximate cell boundaries, and the cell type of all neighboring cells was identified and quantified. Monte Carlo analysis was used to assess whether cell sociology patterns were likely due to random distributions of the cells. Results High density of intra-tumoral CD8+ T cells was significantly associated with non-recurrence of tumors. A cell sociology pattern of CD8+ T cells surrounded by tumor cells was more significantly associated with non-recurrence compared to CD8+ T cell density alone. CD3+ CD8- T cells surrounded by tumor cells was also associated with non-recurrence, but at a similar significance as cell density alone. Cell sociology metrics improved recurrence classifications of 12 patients. Monte Carlo re-sampling analysis determined that these cell sociology patterns were non-random. Conclusion Hyperspectral Cell Sociology expands our understanding of the complex interplay between tumor cells and immune infiltrate. This technology could improve predictions of responses to immunotherapy and lead to a deeper understanding of anti-tumor immunity. Electronic supplementary material The online version of this article (10.1186/s40425-018-0488-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Katey S S Enfield
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada.
| | - Spencer D Martin
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada.,Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Erin A Marshall
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada
| | - Sonia H Y Kung
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada
| | - Paul Gallagher
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada
| | - Katy Milne
- Deeley Research Centre, Victoria, BC, Canada
| | - Zhaoyang Chen
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada
| | - Brad H Nelson
- Deeley Research Centre, Victoria, BC, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Stephen Lam
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada.,Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - John C English
- Pathology and Laboratory Medicine, Vancouver Coastal Health, Vancouver, BC, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Calum E MacAulay
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Wan L Lam
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada.,Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Martial Guillaud
- Integrative Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z1L3, Canada
| |
Collapse
|
154
|
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Department of Dermatology, Yale University School of Medicine, New Haven, CT, United States; Université Paris Descartes/Paris V, Paris, France.
| | - Nils-Petter Rudqvist
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States.
| |
Collapse
|
155
|
Zhou Y, Chen C, Zhang X, Fu S, Xue C, Ma Y, Fang W, Yang Y, Hou X, Huang Y, Zhao H, Hong S, Zhang L. Immune-checkpoint inhibitor plus chemotherapy versus conventional chemotherapy for first-line treatment in advanced non-small cell lung carcinoma: a systematic review and meta-analysis. J Immunother Cancer 2018; 6:155. [PMID: 30577837 PMCID: PMC6303974 DOI: 10.1186/s40425-018-0477-9] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 12/06/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Immune-checkpoint inhibitors plus chemotherapy are emerging as effective first-line treatment in advanced non-small-cell lung carcinoma (NSCLC), but little is known about the magnitude of benefits and potential clinical predictors. METHODS We performed a meta-analysis of randomized trials that compared PD-1/PD-L1 inhibitor plus chemotherapy with chemotherapy in first line of treatment for advanced NSCLC. The outcomes included progression-free survival (PFS), overall survival (OS), objective response rate (ORR) and treatment-related adverse events (AEs). A fixed-effect or random-effects model was adopted depending on between-study heterogeneity. RESULTS Six trials involving 3144 patients were included. PD-1/PD-L1 inhibitor plus chemotherapy was significantly associated with improvement of PFS (hazards ratio [HR], 0.62; 95% CI 0.57-0.67; P < .001), OS (HR, 0.68; 95% CI 0.53-0.87; P = .002) and ORR (relative ratio [RR], 1.56; 95% CI 1.29-1.89; P < .001), irrespective of PD-L1 expression level. The significant predictor(s) for treatment benefit with combination therapy versus chemotherapy alone were PD-L1 expression level for PFS (P < .001); types of checkpoint inhibitor for ORR (P < .001); histology (P = .025), age (P = .038), gender (P < .001), and types of checkpoint inhibitor (P < .001) for OS. In safety analyses, PD-1/PD-L1 inhibitor plus chemotherapy had significantly higher incidence of adverse events (AEs) of grade 3 or higher (RR, 1.14; P = .007), AEs leading to treatment discontinuation (RR, 1.29; P = .022), serious AEs (RR 1.70; P = .006), immune mediated AEs of any grade (RR, 2.37; P < .001), and immune mediated AEs of grade 3 or higher (RR, 3.71; P < .001). CONCLUSIONS PD-1/PD-L1 inhibitor plus chemotherapy, compared with chemotherapy, is associated with significantly improved PFS, ORR, and OS in first-line therapy in NSCLC, at the expense of increased treatment-related AEs.
Collapse
Affiliation(s)
- Yixin Zhou
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of VIP region, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Chen Chen
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Radiotherapy, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xuanye Zhang
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Sha Fu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Pathology Department, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Cong Xue
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Yuxiang Ma
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Wenfeng Fang
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Yunpeng Yang
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Xue Hou
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Yan Huang
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Hongyun Zhao
- State Key Laboratory of Oncology in South China, Guangzhou, China
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Shaodong Hong
- State Key Laboratory of Oncology in South China, Guangzhou, China.
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
| | - Li Zhang
- State Key Laboratory of Oncology in South China, Guangzhou, China.
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
| |
Collapse
|
156
|
Talhouk A, Derocher H, Schmidt P, Leung S, Milne K, Gilks CB, Anglesio MS, Nelson BH, McAlpine JN. Molecular Subtype Not Immune Response Drives Outcomes in Endometrial Carcinoma. Clin Cancer Res 2018; 25:2537-2548. [DOI: 10.1158/1078-0432.ccr-18-3241] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 11/27/2018] [Accepted: 12/03/2018] [Indexed: 11/16/2022]
|
157
|
Santoni M, Conti A, Buti S, Bersanelli M, Foghini L, Piva F, Giulietti M, Lusuardi L, Battelli N. Risk of fatigue in cancer patients treated with anti programmed cell death-1/anti programmed cell death ligand-1 agents: a systematic review and meta-analysis. Immunotherapy 2018; 10:1303-1313. [PMID: 30474475 DOI: 10.2217/imt-2018-0067] [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 We aimed to assess the incidence and relative risk (RR) of fatigue in cancer patients treated with anti programmed cell death-1 (PD-1) and anti programmed cell death ligand-1 (PD-L1) agents. PATIENTS & METHODS Eligible studies were selected according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. Incidence, RR and 95% CIs were calculated using random or fixed-effects models. RESULTS Thirty-eight studies were included in this analysis, with a total of 11,719 patients. The incidences were 23.4 and 2.1% for all- and high-grade fatigue, respectively. The highest incidence of high-grade fatigue was reported by the combination of nivolumab and ipilimumab. Overall RR of high-grade fatigue with anti-PD-1/PD-L1 compared with chemotherapy or targeted therapy was 0.48. CONCLUSION Treatment with anti-PD-1/PD-L1 agents correlates with lower incidence and RR of fatigue compared with standard therapies.
Collapse
Affiliation(s)
- Matteo Santoni
- Oncology Unit, Macerata Hospital, via Santa Lucia 2, 62100, Macerata, Italy
| | - Alessandro Conti
- Azienda Ospedaliera dell'Alto Adige, Bressanone/Brixen Hospital, Via Dante, 51, 39042, Italy
| | - Sebastiano Buti
- University Hospital of Parma, Via Gramsci 14 - 43126, Parma, Italy
| | | | - Laura Foghini
- Oncology Unit, Macerata Hospital, via Santa Lucia 2, 62100, Macerata, Italy
| | - Francesco Piva
- Department of Specialistic Clinical & Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Matteo Giulietti
- Department of Specialistic Clinical & Odontostomatological Sciences, Polytechnic University of Marche, Ancona, Italy
| | - Lukas Lusuardi
- Department of Urology & Andrology, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Nicola Battelli
- Oncology Unit, Macerata Hospital, via Santa Lucia 2, 62100, Macerata, Italy
| |
Collapse
|
158
|
Radpour R, Forouharkhou F. Single-cell analysis of tumors: Creating new value for molecular biomarker discovery of cancer stem cells and tumor-infiltrating immune cells. World J Stem Cells 2018; 10:160-171. [PMID: 30631391 PMCID: PMC6325074 DOI: 10.4252/wjsc.v10.i11.160] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 10/18/2018] [Accepted: 10/23/2018] [Indexed: 02/06/2023] Open
Abstract
Biomarker-driven individualized treatment in oncology has made tremendous progress through technological developments, new therapeutic modalities and a deeper understanding of the molecular biology for tumors, cancer stem cells and tumor-infiltrating immune cells. Recent technical developments have led to the establishment of a variety of cancer-related diagnostic, prognostic and predictive biomarkers. In this regard, different modern OMICs approaches were assessed in order to categorize and classify prognostically different forms of neoplasia. Despite those technical advancements, the extent of molecular heterogeneity at the individual cell level in human tumors remains largely uncharacterized. Each tumor consists of a mixture of heterogeneous cell types. Therefore, it is important to quantify the dynamic cellular variations in order to predict clinical parameters, such as a response to treatment and or potential for disease recurrence. Recently, single-cell based methods have been developed to characterize the heterogeneity in seemingly homogenous cancer cell populations prior to and during treatment. In this review, we highlight the recent advances for single-cell analysis and discuss the challenges and prospects for molecular characterization of cancer cells, cancer stem cells and tumor-infiltrating immune cells.
Collapse
Affiliation(s)
- Ramin Radpour
- Tumor Immunology, Department for BioMedical Research (DBMR), University of Bern, Bern 3008, Switzerland
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern 3008, Switzerland
| | - Farzad Forouharkhou
- Department for Bioinformatics, Persian Bioinformatics System, Tehran 14166, Iran
| |
Collapse
|
159
|
Menard LC, Fischer P, Kakrecha B, Linsley PS, Wambre E, Liu MC, Rust BJ, Lee D, Penhallow B, Manjarrez Orduno N, Nadler SG. Renal Cell Carcinoma (RCC) Tumors Display Large Expansion of Double Positive (DP) CD4+CD8+ T Cells With Expression of Exhaustion Markers. Front Immunol 2018; 9:2728. [PMID: 30534127 PMCID: PMC6275222 DOI: 10.3389/fimmu.2018.02728] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 11/05/2018] [Indexed: 02/01/2023] Open
Abstract
Checkpoint inhibitors target the inhibitory receptors expressed by tumor-infiltrating T cells in order to reinvigorate an anti-tumor immune response. Therefore, understanding T cell composition and phenotype in human tumors is crucial. We analyzed by flow cytometry tumor-infiltrating lymphocytes (TILs) from two independent cohorts of patients with different cancer types, including RCC, lung, and colon cancer. In healthy donors, peripheral T cells are usually either CD4+ or CD8+ with a small percentage of CD4+ CD8+ DP cells (<5%). Compared to several other cancer types, including lung, and colorectal cancers, TILs from about a third of RCC patients showed an increased proportion of DP CD4+CD8+ T cells (>5%, reaching 30–50% of T cells in some patients). These DP T cells have an effector memory phenotype and express CD38, 4-1BB, and HLA-DR, suggesting antigen-driven expansion. In fact, TCR sequencing analysis revealed a high degree of clonality in DP T cells. Additionally, there were high levels of PD-1 and TIM-3 expression on DP T cells, which correlated with higher expression of PD-1 and TIM-3 in conventional single positive CD8 T cells from the same patients. These results suggest that DP T cells could be dysfunctional tumor-specific T cells with the potential to be reactivated by checkpoint inhibitors.
Collapse
Affiliation(s)
- Laurence C Menard
- Translational Medicine, Bristol-Myers Squibb, Princeton, NJ, United States
| | - Paul Fischer
- Translational Medicine, Bristol-Myers Squibb, Princeton, NJ, United States
| | - Bijal Kakrecha
- Translational Medicine, Bristol-Myers Squibb, Princeton, NJ, United States
| | - Peter S Linsley
- Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Erik Wambre
- Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Maochang C Liu
- Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Blake J Rust
- Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - Deborah Lee
- Translational Medicine, Bristol-Myers Squibb, Princeton, NJ, United States
| | - Becky Penhallow
- Translational Medicine, Bristol-Myers Squibb, Princeton, NJ, United States
| | | | - Steven G Nadler
- Translational Medicine, Bristol-Myers Squibb, Princeton, NJ, United States
| |
Collapse
|
160
|
Ma LJ, Feng FL, Dong LQ, Zhang Z, Duan M, Liu LZ, Shi JY, Yang LX, Wang ZC, Zhang S, Ding ZB, Ke AW, Cao Y, Zhang XM, Zhou J, Fan J, Wang XY, Gao Q. Clinical significance of PD-1/PD-Ls gene amplification and overexpression in patients with hepatocellular carcinoma. Theranostics 2018; 8:5690-5702. [PMID: 30555574 PMCID: PMC6276293 DOI: 10.7150/thno.28742] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 10/04/2018] [Indexed: 12/14/2022] Open
Abstract
Background: The remarkable clinical activity of PD-1 antibody in advanced hepatocellular carcinoma (HCC) highlights the importance of PD-1/PD-L1-mediated immune escape as therapeutic target in HCC. However, the frequency and prognostic significance of PD-Ls genetic alterations in HCC remain unknown. Methods: Fluorescence in situ hybridization were used to determine PD-Ls genetic alterations, and qPCR data coupled with immunofluorescence were used to measure the mRNA and protein levels of PD-Ls. Clinical relevance and prognostic value of 9p24.1 genetic alterations were investigated on tissue microarray containing three independent cohorts of 578 HCC patients. The results were further validated in an independent cohort of 442 HCC patients from The Cancer Genome Atlas (TCGA) database. Results: In total, 7.1%-15.0% for amplification and 15.8%-31.3% for polysomy of 9p24.1 were revealed in three cohorts of HCC patients, similar to the objective response rate of PD-1 antibody in HCC. Patients with 9p24.1 genetic alterations significantly and independently correlated with unfavorable outcomes than those without. FISH and qPCR data coupled with immunofluorescence revealed that genetic alterations of 9p24.1 robustly contributed to PD-L1 and PD-L2 upregulation. In addition, increased expression of PD-L1 instead of PD-L2 also predicted poor survival by multivariate analyses. Meanwhile, high infiltration of PD-1+ immune cells also indicated dismal survival in HCC. Conclusions: Amplification or higher expression of PD-L1 significantly and independently correlated with unfavorable survival in HCC patients, authenticating the PD-1/PD-L1 axis as rational immunotherapeutic targets for HCC.
Collapse
Affiliation(s)
- Li-Jie Ma
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
| | - Fei-Ling Feng
- Department of Biliary Surgery, Eastern Hepatobiliary Surgical Hospital, Naval Military Medical University, Shanghai 200082, China
| | - Liang-Qing Dong
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
| | - Zhao Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
| | - Meng Duan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
| | - Long-Zi Liu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
| | - Jie-Yi Shi
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
| | - Liu-Xiao Yang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
| | - Zhi-Chao Wang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
| | - Shu Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
| | - Zhen-Bin Ding
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
| | - Ai-Wu Ke
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
| | - Ya Cao
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China
| | - Xiao-Ming Zhang
- Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200032, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiao-Ying Wang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Shanghai, China
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai 200433, China
| |
Collapse
|
161
|
Villaruz LC, Ancevski Hunter K, Kurland BF, Abberbock S, Herbst C, Dacic S. Comparison of PD-L1 immunohistochemistry assays and response to PD-1/L1 inhibitors in advanced non-small-cell lung cancer in clinical practice. Histopathology 2018; 74:269-275. [PMID: 30105871 DOI: 10.1111/his.13729] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 08/11/2018] [Indexed: 01/05/2023]
Abstract
AIMS Several studies have demonstrated analytical comparability between different PD-L1 assays, but their clinical validity in non-small-cell lung cancer in terms of response to treatment outside clinical trials has not been established. The aim of our study is to assess the analytical performance of laboratory-developed tests for Ventana SP263 and Agilent/Dako 22C3, and to investigate the association between PD-L1 assays and response to PD-1/L1 inhibitors. METHODS AND RESULTS PD-L1 SP263 and 22C3 assays were performed on 302 consecutive non-small-cell lung carcinoma samples Both assays were optimised for use on the automated Ventana BenchMark Ultra platform. Scoring algorithms for staining of the tumour cells using the established cut-offs were applied to all samples. Best overall response (BOR) for 44 patients treated with either nivolumab, pembrolizumab or atezolizumab were assessed using recist version 1.1 and correlated with PD-L1 assay results. Assays showed good agreement, with a concordance correlation coefficient of 0.86 [95% confidence interval (CI) = 0.82-0.90)]. Comparing the assays using cut-offs of 1%, 5%, 10%, 1-49% and ≥50% showed an association between the two assays (P < 0.0001). The SP263 10% cut-off (P = 0.032) was associated with BOR, whereas the 1% (P = 0.087) and 5% (P = 0.051) cut-offs were not. In contrast 22C3, cut-offs of 1% (P = 0.019), 5% (P = 0.025) and 10% (P = 0.014) were all associated with BOR. CONCLUSIONS The SP263 and 22C3 LDTs demonstrated good analytical concordance, and correlation with response to PD-1/L1 inhibitors.
Collapse
Affiliation(s)
| | | | - Brenda F Kurland
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Biostatistics, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Cameron Herbst
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Sanja Dacic
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA.,Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
162
|
Yu H, Chen Z, Ballman KV, Watson MA, Govindan R, Lanc I, Beer DG, Bueno R, Chirieac LR, Chui MH, Chen G, Franklin WA, Gandara DR, Genova C, Brovsky KA, Joshi MBM, Merrick DT, Richards WG, Rivard CJ, Harpole DH, Tsao MS, van Bokhoven A, Shepherd FA, Hirsch FR. Correlation of PD-L1 Expression with Tumor Mutation Burden and Gene Signatures for Prognosis in Early-Stage Squamous Cell Lung Carcinoma. J Thorac Oncol 2018; 14:25-36. [PMID: 30253973 DOI: 10.1016/j.jtho.2018.09.006] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 07/13/2018] [Accepted: 09/10/2018] [Indexed: 02/06/2023]
Abstract
OBJECTIVES Anti-programmed cell death 1 (PD-1)/programmed death ligand 1 (PD-L1) immunotherapy has demonstrated success in the treatment of advanced NSCLC. Recently, PD-1/PD-L1 blockade also has demonstrated interesting results in small trials of neoadjuvant treatment in stage IB to IIIA NSCLC. In addition, several clinical trials using anti-PD-1/PD-L1 immunotherapy as an adjuvant or neoadjuvant treatment in patients with resectable stage NSCLC are ongoing. However, few analyses of anti-PD-1/PD-L1 immunotherapy-related biomarkers in early-stage squamous cell lung carcinoma (SqCLC) have been reported. In this study, we evaluated PD-L1 protein expression, tumor mutation burden, and expression of an immune gene signature in early-stage SqCLC, providing data for identifying the potential role for patients with anti-PD-1/PD-L1 treatment in early-stage SqCLC. METHODS A total of 255 specimens from patients with early-stage SqCLC were identified within participating centers of the Strategic Partnering to Evaluate Cancer Signatures program. PD-L1 protein expression by immunohistochemistry was evaluated by using the Dako PD-L1 22C3 pharmDx kit on the Dako Link 48 auto-stainer (Dako, Carpinteria, CA). Tumor mutation burden (TMB) was calculated on the basis of data from targeted genome sequencing. The T-effector and interferon gamma (IFN-γ) gene signature was determined from Affymetrix gene chip data (Affymetrix, Santa Clara, CA) from frozen specimens. RESULTS The prevalence of PD-L1 expression was 9.8% at a tumor proportion score cutoff of at least 50%. PD-L1 mRNA and programmed cell death 1 ligand 2 mRNA positively correlated with PD-L1 protein expression on tumor cells (TCs) and tumor-infiltrating immune cells. PD-L1 protein expression on tumor-infiltrating immune cells was correlated with the T-effector and IFN-γ gene signature (p < 0.001), but not with TMB. For TCs, all of these biomarkers were independent of each other and neither PD-L1 protein expression, TMB, or T-effector and IFN-γ gene signatures were independently prognostic for patient outcomes. CONCLUSIONS Evaluation of PD-L1 expression, TMB, and T-effector and IFN-γ gene signatures in the cohort with early-stage SqCLC found them to be independent of each other, and none was associated with overall survival. Our results also support the hypothesis that PD-L1 expression is regulated by an intrinsic mechanism on TCs and an adaptive mechanism on immune cells.
Collapse
Affiliation(s)
- Hui Yu
- Division of Medical Oncology, University of Colorado Denver, Aurora, Colorado
| | - Zhengming Chen
- Division of Biostatistics and Epidemiology, Department of Healthcare Policy and Research, Weill Cornell Medicine, New York, New York
| | - Karla V Ballman
- Division of Biostatistics and Epidemiology, Department of Healthcare Policy and Research, Weill Cornell Medicine, New York, New York
| | - Mark A Watson
- Washington University School of Medicine, St. Louis, Missouri
| | | | - Irena Lanc
- Washington University School of Medicine, St. Louis, Missouri
| | | | - Raphael Bueno
- Brigham and Women's Hospital/Harvard Medical School, Boston, Massachusetts
| | | | - Michael Herman Chui
- University Health Network/Princess Margaret Cancer Centre and University of Toronto, Toronto, Canada
| | - Guoan Chen
- University of Michigan, Ann Arbor, Michigan
| | - Wilbur A Franklin
- Department of Pathology, University of Colorado Denver, Aurora, Colorado
| | - David R Gandara
- University of California Davis Comprehensive Cancer Center, Sacramento, California
| | - Carlo Genova
- Lung Cancer Unit, San Martino Hospital, Genoa, Italy
| | - Kristine A Brovsky
- Division of Medical Oncology, University of Colorado Denver, Aurora, Colorado
| | | | - Daniel T Merrick
- Department of Pathology, University of Colorado Denver, Aurora, Colorado
| | - William G Richards
- Brigham and Women's Hospital/Harvard Medical School, Boston, Massachusetts
| | | | | | - Ming-Sound Tsao
- University Health Network/Princess Margaret Cancer Centre and University of Toronto, Toronto, Canada
| | - Adrie van Bokhoven
- Department of Pathology, University of Colorado Denver, Aurora, Colorado
| | - Frances A Shepherd
- University Health Network/Princess Margaret Cancer Centre and University of Toronto, Toronto, Canada
| | - Fred R Hirsch
- Division of Medical Oncology, University of Colorado Denver, Aurora, Colorado.
| |
Collapse
|
163
|
Ninomiya K, Hotta K. Pembrolizumab for the first-line treatment of non-small cell lung cancer. Expert Opin Biol Ther 2018; 18:1015-1021. [PMID: 30207786 DOI: 10.1080/14712598.2018.1522300] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
INTRODUCTION Platinum-based chemotherapy had long played a role as standard therapy for the first-line treatment of advanced or recurrent non-small cell lung cancer (NSCLC). However, immune checkpoint inhibitors such as pembrolizumab, a monoclonal antibody that prevents programmed death protein 1 (PD-1) receptor, have brought a paradigm shift in this field. Areas covered: In this article, we review the relevant literatures and ongoing trials on the first-line treatment of pembrolizumab. Especially, in two pivotal phase III trials, KEYNOTE-024 and -189, both pembrolizumab monotherapy and combined pembrolizumab plus chemotherapy significantly prolonged overall survival (OS) compared to the existing platinum-based chemotherapy. Currently, multiple trials with combination therapy of pembrolizumab and other agents have been conducted, and further evidences are expected to be created. Expert opinion: Immune checkpoint inhibitors that block the PD-1/PD-L1 pathway are essential drugs for advanced or recurrent NSCLC, among which pembrolizumab becomes one of the standards of care in the first-line of NSCLC. For further improvement in efficacy of pembrolizumab, it is necessary to clarify the identification of biomarkers exclusive to PD-L1 expression, predictive factors for patients who benefit most from the agent.
Collapse
Affiliation(s)
- Kiichiro Ninomiya
- a Department of Hematology, Oncology and Respiratory Medicine , Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences , Okayama , Japan
| | - Katsuyuki Hotta
- b Center of Innovative Clinical Medicine , Okayama University Hospital , Okayama , Japan
| |
Collapse
|
164
|
Hsu JC, Lin JY, Hsu MY, Lin PC. Effectiveness and safety of immune checkpoint inhibitors: A retrospective study in Taiwan. PLoS One 2018; 13:e0202725. [PMID: 30142174 PMCID: PMC6108484 DOI: 10.1371/journal.pone.0202725] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 08/08/2018] [Indexed: 12/13/2022] Open
Abstract
Background Since 2012, several immune checkpoint inhibitors have been approved by the Taiwan FDA for various types of cancer treatment. However, none of them are covered by Taiwan National Health Insurance due to the fact that they are expensive, and there is a lack of clinical evidence as to their effectiveness. Objectives This study was aimed toward an exploration of clinical experiences with use of immune checkpoint inhibitors, including indications, prescription types, drug effectiveness, adverse drug event types, and incidence, all of which shall serve as references for future clinical drug use. Methods This is a retrospective study focusing on three immune checkpoint inhibitors (ipilimumab, nivolumab, and pembrolizumab), which are available for cancer treatment in Taiwan. We collected data from medical records for the period from January 1st, 2015 to January 12th, 2017 at National Cheng Kung University Hospital (NCKUH), a medical center in southern Taiwan, and recorded these cases until May 31st, 2017. Overall survival (OS) and progression-free survival (PFS) were estimated using the Kaplan-Meier method, and adverse drug reaction odds ratios were analyzed using a chi-square analysis. Results The 50 patients under consideration in this study had used any one of the immune checkpoint inhibitors in NCKUH. Non-small cell lung cancer (n = 24, 48%) accounted for the highest percentage, followed by hepatocellular carcinoma (n = 4, 8%). The median OS was not reached, and the PFS for all immunotherapies was 4.9 months. The median OS period and PFS for non-small cell lung cancer (NSCLC) patients were 13 and 4.9 months, respectively, which were similar to those in many clinical trials. For NSCLC patients, the OS and PFS were only 0.63 and 1.37 months for squamous cell type NSCLC, and for patients who were PD-L1 negative, the OS and PFS were only 11.53 and 2.6 months, respectively. The most common adverse events in this study included fatigue (42%), rashes (22%), nausea (20%), and fever (20%), while one patient developed severe deep venous thrombosis and tissue inflammation, which was not confirmed in previous clinical trials. Conclusions The histological subtype, the intensity of the PD-L1 expression, and the timing of treatment affected the NSCLC therapeutic results. It is recommended that clinical tests be conducted in order to enhance therapeutic effectiveness. It is expected that more testing, observation-based studies, and research results will validate their efficacy and the tolerance levels of patients.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Antibodies, Monoclonal, Humanized/adverse effects
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/therapeutic use
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Non-Small-Cell Lung/pathology
- Disease-Free Survival
- Fatigue/etiology
- Female
- Humans
- Immunotherapy
- Ipilimumab/adverse effects
- Ipilimumab/therapeutic use
- Kaplan-Meier Estimate
- Lung Neoplasms/drug therapy
- Lung Neoplasms/mortality
- Lung Neoplasms/pathology
- Male
- Middle Aged
- Nivolumab/adverse effects
- Nivolumab/therapeutic use
- Retrospective Studies
- Taiwan
- Treatment Outcome
Collapse
Affiliation(s)
- Jason C. Hsu
- School of Pharmacy and Institute of Clinical Pharmacy and Pharmaceutical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- * E-mail:
| | - Jia-Yu Lin
- Department of Pharmacy, National Cheng Kung University Hospital, Tainan, Taiwan
| | - May-Ying Hsu
- Department of Pharmacy, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Peng-Chan Lin
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
| |
Collapse
|
165
|
Yi M, Jiao D, Xu H, Liu Q, Zhao W, Han X, Wu K. Biomarkers for predicting efficacy of PD-1/PD-L1 inhibitors. Mol Cancer 2018; 17:129. [PMID: 30139382 PMCID: PMC6107958 DOI: 10.1186/s12943-018-0864-3] [Citation(s) in RCA: 560] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 07/24/2018] [Indexed: 12/30/2022] Open
Abstract
Programmed cell death protein 1/programmed cell death ligand 1 (PD-1/PD-L1) is a negative modulatory signaling pathway for activation of T cell. It is acknowledged that PD-1/PD-L1 axis plays a crucial role in the progression of tumor by altering status of immune surveillance. As one of the most promising immune therapy strategies, PD-1/PD-L1 inhibitor is a breakthrough for the therapy of some refractory tumors. However, response rate of PD-1/PD-L1 inhibitors in overall patients is unsatisfactory, which limits the application in clinical practice. Therefore, biomarkers which could effectively predict the efficacy of PD-1/PD-L1 inhibitors are crucial for patient selection. Biomarkers reflecting tumor immune microenvironment and tumor cell intrinsic features, such as PD-L1 expression, density of tumor infiltrating lymphocyte (TIL), tumor mutational burden, and mismatch-repair (MMR) deficiency, have been noticed to associate with treatment effect of anti-PD-1/anti-PD-L1 therapy. Furthermore, gut microbiota, circulating biomarkers, and patient previous history have been found as valuable predictors as well. Therefore establishing a comprehensive assessment framework involving multiple biomarkers would be meaningful to interrogate tumor immune landscape and select sensitive patients.
Collapse
Affiliation(s)
- Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Dechao Jiao
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hanxiao Xu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Qian Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weiheng Zhao
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
166
|
Blood-based tumor mutational burden as a predictor of clinical benefit in non-small-cell lung cancer patients treated with atezolizumab. Nat Med 2018; 24:1441-1448. [PMID: 30082870 DOI: 10.1038/s41591-018-0134-3] [Citation(s) in RCA: 876] [Impact Index Per Article: 125.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/22/2018] [Indexed: 02/08/2023]
Abstract
Although programmed death-ligand 1-programmed death 1 (PD-L1-PD-1) inhibitors are broadly efficacious, improved outcomes have been observed in patients with high PD-L1 expression or high tumor mutational burden (TMB). PD-L1 testing is required for checkpoint inhibitor monotherapy in front-line non-small-cell lung cancer (NSCLC). However, obtaining adequate tumor tissue for molecular testing in patients with advanced disease can be challenging. Thus, an unmet medical need exists for diagnostic approaches that do not require tissue to identify patients who may benefit from immunotherapy. Here, we describe a novel, technically robust, blood-based assay to measure TMB in plasma (bTMB) that is distinct from tissue-based approaches. Using a retrospective analysis of two large randomized trials as test and validation studies, we show that bTMB reproducibly identifies patients who derive clinically significant improvements in progression-free survival from atezolizumab (an anti-PD-L1) in second-line and higher NSCLC. Collectively, our data show that high bTMB is a clinically actionable biomarker for atezolizumab in NSCLC.
Collapse
|
167
|
Manjarrez-Orduño N, Menard LC, Kansal S, Fischer P, Kakrecha B, Jiang C, Cunningham M, Greenawalt D, Patel V, Yang M, Golhar R, Carman JA, Lezhnin S, Dai H, Kayne PS, Suchard SJ, Bernstein SH, Nadler SG. Circulating T Cell Subpopulations Correlate With Immune Responses at the Tumor Site and Clinical Response to PD1 Inhibition in Non-Small Cell Lung Cancer. Front Immunol 2018; 9:1613. [PMID: 30123214 PMCID: PMC6085412 DOI: 10.3389/fimmu.2018.01613] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 06/28/2018] [Indexed: 01/13/2023] Open
Abstract
Agents targeting the PD1-PDL1 axis have transformed cancer therapy. Factors that influence clinical response to PD1-PDL1 inhibitors include tumor mutational burden, immune infiltration of the tumor, and local PDL1 expression. To identify peripheral correlates of the anti-tumor immune response in the absence of checkpoint blockade, we performed a retrospective study of circulating T cell subpopulations and matched tumor gene expression in melanoma and non-small cell lung cancer (NSCLC) patients. Notably, both melanoma and NSCLC patients whose tumors exhibited increased inflammatory gene transcripts presented high CD4+ and CD8+ central memory T cell (CM) to effector T cell (Eff) ratios in blood. Consequently, we evaluated CM/Eff T cell ratios in a second cohort of NSCLC. The data showed that high CM/Eff T cell ratios correlated with increased tumor PDL1 expression. Furthermore, of the 22 patients within this NSCLC cohort who received nivolumab, those with high CM/Eff T cell ratios, had longer progression-free survival (PFS) (median survival: 91 vs. 215 days). These findings show that by providing a window into the state of the immune system, peripheral T cell subpopulations inform about the state of the anti-tumor immune response and identify potential blood biomarkers of clinical response to checkpoint inhibitors in melanoma and NSCLC.
Collapse
Affiliation(s)
| | | | | | - Paul Fischer
- Bristol-Myers Squibb, Princeton, NJ, United States
| | | | - Can Jiang
- Bristol-Myers Squibb, Princeton, NJ, United States
| | | | | | - Vishal Patel
- Bristol-Myers Squibb, Princeton, NJ, United States
| | - Minghui Yang
- Bristol-Myers Squibb, Princeton, NJ, United States
| | - Ryan Golhar
- Bristol-Myers Squibb, Princeton, NJ, United States
| | | | | | | | - Paul S Kayne
- Bristol-Myers Squibb, Princeton, NJ, United States
| | | | | | | |
Collapse
|
168
|
Khan M, Lin J, Liao G, Tian Y, Liang Y, Li R, Liu M, Yuan Y. Comparative analysis of immune checkpoint inhibitors and chemotherapy in the treatment of advanced non-small cell lung cancer: A meta-analysis of randomized controlled trials. Medicine (Baltimore) 2018; 97:e11936. [PMID: 30113497 PMCID: PMC6113026 DOI: 10.1097/md.0000000000011936] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Recently, immune checkpoint inhibitors have shown survival advantage over chemotherapy in the treatment of advanced non-small cell lung cancer (NSCLC). This meta-analysis was conducted to gather and analyze the available evidence (Evidence level I; Randomized Controlled Trials) comparing efficacy and safety of anti-programmed cell death-1 (PD1)/programmed cell death ligand 1 (PD-L1) therapies and chemotherapy in the treatment of advanced NSCLC. METHODS A search strategy was devised to identify the randomized controlled trials (RCTs) using electronic databases of PubMed, Cochrane Library, and Web of Science. Hazard ratios or odds ratios obtained for overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and treatment related adverse events (TRAEs) were analyzed using fixed effect model or random effects model. Additionally, subgroup analysis was also performed. RESULTS A total of seven RCTs (n = 3867) were identified and selected for inclusion in this meta-analysis. Anti-PD1/PD-L1 therapies (nivolumab, pembrolizumab, atezolizumab) resulted in better OS (HR 0.72 [95% confidence interval [CI] 0.63, 0.82; P < .00001]), PFS (HR 0.84 [95% CI 0.72, 0.97; P < .02]), and ORR (odds ratio [OR] 1.52 [95% CI 1.08, 2.14; P < .02]) in comparison to chemotherapy in advanced NSCLC. Improved safety was observed with anti-PD1/PD-L1 therapies (OR 0.31 [95%CI 0.26, 0.38; P < .00001]). Subgroups analysis revealed Eastern Cooperative Oncology Group Performance Status (ECOG PS) 1 (HR 0.76 [95%CI 0.62, 0.93; P = .007]), squamous cell type (HR 0.76 [95% CI 0.63, 0.92; P = .005]), current/former smoker (HR 0.76 [95% CI 0.63, 0.92; P = .005]), epidermal growth factor receptor (EGFR) wild type (HR 0.67 [95% CI 0.60, 0.76; P < .00001]), Kirsten rat sarcoma oncogene mutation (KRAS) mutant (HR 0.60 [95% CI 0.39, 0.93; P < .02]), and absence of central nervous system (CNS) metastases (HR 0.71 [95% CI 0.63, 0.80; P < .00001]) were associated with better overall survival. CONCLUSIONS Anti-PD1/PD-L1 therapies are safe and effective treatment option in advanced non-small cell lung cancer and can be recommended selectively.
Collapse
Affiliation(s)
- Muhammad Khan
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University
| | - Jie Lin
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University
| | - Guixiang Liao
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University
| | - Yunhong Tian
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University
| | - Yingying Liang
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University
| | - Rong Li
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University
| | - Mengzhong Liu
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Sun Yat-sen Medical University, Guangzhou, Guangdong Province, People's Republic of China
| | - Yawei Yuan
- Department of Radiation Oncology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, Sun Yat-sen Medical University, Guangzhou, Guangdong Province, People's Republic of China
| |
Collapse
|
169
|
Meyers D, Bryan P, Banerji S, Morris D. Targeting the PD-1/PD-L1 axis for the treatment of non-small-cell lung cancer. Curr Oncol 2018; 25:e324-e334. [PMID: 30111979 PMCID: PMC6092051 DOI: 10.3747/co.25.3976] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Lung cancer is the leading cause of cancer-specific death among Canadians, with non-small-cell lung cancer (nsclc) being the most common histologic variant. Despite advances in the understanding of the molecular biology of nsclc, the survival rate for this malignancy is still poor. It is now understood that, to evade detection and immune clearance, nsclc tumours overexpress the immunosuppressive checkpoint protein programmed death ligand 1 (PD-L1). Inhibiting the PD-1/PD-L1 axis with monoclonal antibodies has significantly changed the treatment landscape in nsclc during the last 5 years. Despite evidence of clinical response in some patients, only approximately 20% of patients obtain any durable benefit, and many of the patients who do respond ultimately relapse with drug-resistant disease. The identification of patients who are most likely to benefit from such therapy is therefore important. In the present review, we cover the basics of the PD-1/PD-L1 axis and its clinical significance in nsclc, biomarkers that are predictive of treatment response, relevant clinical trials of PD-1/PD-L1 blockade completed to date, and proposed mechanisms of acquired therapeutic resistance.
Collapse
Affiliation(s)
- D.E. Meyers
- Department of Oncology, University of Calgary, and
- Tom Baker Cancer Centre, Calgary, AB
| | - P.M. Bryan
- Department of Oncology, University of Calgary, and
| | - S. Banerji
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, and
- Department of Medical Oncology, CancerCare Manitoba, Winnipeg, MB
| | - D.G. Morris
- Department of Oncology, University of Calgary, and
- Tom Baker Cancer Centre, Calgary, AB
| |
Collapse
|
170
|
Testa U, Castelli G, Pelosi E. Lung Cancers: Molecular Characterization, Clonal Heterogeneity and Evolution, and Cancer Stem Cells. Cancers (Basel) 2018; 10:E248. [PMID: 30060526 PMCID: PMC6116004 DOI: 10.3390/cancers10080248] [Citation(s) in RCA: 251] [Impact Index Per Article: 35.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 07/19/2018] [Accepted: 07/20/2018] [Indexed: 12/21/2022] Open
Abstract
Lung cancer causes the largest number of cancer-related deaths in the world. Most (85%) of lung cancers are classified as non-small-cell lung cancer (NSCLC) and small-cell lung cancer (15%) (SCLC). The 5-year survival rate for NSCLC patients remains very low (about 16% at 5 years). The two predominant NSCLC histological phenotypes are adenocarcinoma (ADC) and squamous cell carcinoma (LSQCC). ADCs display several recurrent genetic alterations, including: KRAS, BRAF and EGFR mutations; recurrent mutations and amplifications of several oncogenes, including ERBB2, MET, FGFR1 and FGFR2; fusion oncogenes involving ALK, ROS1, Neuregulin1 (NRG1) and RET. In LSQCC recurrent mutations of TP53, FGFR1, FGFR2, FGFR3, DDR2 and genes of the PI3K pathway have been detected, quantitative gene abnormalities of PTEN and CDKN2A. Developments in the characterization of lung cancer molecular abnormalities provided a strong rationale for new therapeutic options and for understanding the mechanisms of drug resistance. However, the complexity of lung cancer genomes is particularly high, as shown by deep-sequencing studies supporting the heterogeneity of lung tumors at cellular level, with sub-clones exhibiting different combinations of mutations. Molecular studies performed on lung tumors during treatment have shown the phenomenon of clonal evolution, thus supporting the occurrence of a temporal tumor heterogeneity.
Collapse
Affiliation(s)
- Ugo Testa
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Germana Castelli
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy.
| | - Elvira Pelosi
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy.
| |
Collapse
|
171
|
Bylicki O, Paleiron N, Rousseau-Bussac G, Chouaïd C. New PDL1 inhibitors for non-small cell lung cancer: focus on pembrolizumab. Onco Targets Ther 2018; 11:4051-4064. [PMID: 30038505 PMCID: PMC6052930 DOI: 10.2147/ott.s154606] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The advent of immune-checkpoint inhibitors during the past decade represents a major advancement in the treatment of non-small cell lung cancer (NSCLC) with personalized treatment. Platinum-based chemotherapy has reached its efficacy threshold, with its use remaining limited by its toxicity. For NSCLC, inhibitors of the PD1 protein and its ligand PDL1 show promising clinical activity and induce durable responses in patients with advanced disease. The US Food and Drug Administration has approved pembrolizumab for treatment-naïve metastatic NSCLC with ≥50% of tumor cells expressing PDL1 and for metastatic NSCLC with ≥1% PDL1 expression after progression following first-line platinum-based doublet chemotherapy. In 2017, it also authorized the first-line combination of pembrolizumab and carboplatin-pemetrexed chemotherapy without selection based on PDL1 expression, but European health authorities are still waiting for the results of a Phase III trial. In this review, the clinical results of published and ongoing studies evaluating pembrolizumab for advanced NSCLC are analyzed and the potential role of PDL1 as a factor predictive of overall responses addressed.
Collapse
Affiliation(s)
- Olivier Bylicki
- Pneumology Department, Hôpital d'Instruction des Armées Percy, Paris,
| | - Nicolas Paleiron
- Respiratory Department, Hôpital d'Instruction des Armées Saint-Anne, Toulon
| | | | | |
Collapse
|
172
|
Saigi M, Alburquerque-Bejar JJ, Mc Leer-Florin A, Pereira C, Pros E, Romero OA, Baixeras N, Esteve-Codina A, Nadal E, Brambilla E, Sanchez-Cespedes M. MET-Oncogenic and JAK2-Inactivating Alterations Are Independent Factors That Affect Regulation of PD-L1 Expression in Lung Cancer. Clin Cancer Res 2018; 24:4579-4587. [PMID: 29898990 DOI: 10.1158/1078-0432.ccr-18-0267] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 05/10/2018] [Accepted: 06/06/2018] [Indexed: 11/16/2022]
Abstract
Purpose: The blockade of immune checkpoints such as PD-L1 and PD-1 is being exploited therapeutically in several types of malignancies. Here, we aimed to understand the contribution of the genetics of lung cancer to the ability of tumor cells to escape immunosurveillance checkpoints.Experimental Design: More than 150 primary non-small cell lung cancers, including pulmonary sarcomatoid carcinomas, were tested for levels of the HLA-I complex, PD-L1, tumor-infiltrating CD8+ lymphocytes, and alterations in main lung cancer genes. Correlations were validated in cancer cell lines using appropriate treatments to activate or inhibit selected pathways. We also performed RNA sequencing to assess changes in gene expression after these treatments.Results:MET-oncogenic activation tended to associate with positive PD-L1 immunostaining, whereas STK11 mutations were correlated with negative immunostaining. In MET-altered cancer cells, MET triggered a transcriptional increase of PD-L1 that was independent of the IFNγ-mediated JAK/STAT pathway. The activation of MET also upregulated other immunosuppressive genes (PDCD1LG2 and SOCS1) and transcripts involved in angiogenesis (VEGFA and NRP1) and in cell proliferation. We also report recurrent inactivating mutations in JAK2 that co-occur with alterations in MET and STK11, which prevented the induction of immunoresponse-related genes following treatment with IFNγ.Conclusions: We show that MET activation promotes the expression of several negative checkpoint regulators of the immunoresponse, including PD-L1. In addition, we report inactivation of JAK2 in lung cancer cells that prevented the response to IFNγ. These alterations are likely to facilitate tumor growth by enabling immune tolerance and may affect the response to immune checkpoint inhibitors. Clin Cancer Res; 24(18); 4579-87. ©2018 AACR.
Collapse
Affiliation(s)
- Maria Saigi
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Juan J Alburquerque-Bejar
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Anne Mc Leer-Florin
- Département d'Anatomie et Cytologie Pathologiques, Pôle de Biologie et Pathologie, CHU Grenoble Alpes, Grenoble and Université Grenoble Alpes, Grenoble, France
| | - Carolina Pereira
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Eva Pros
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Octavio A Romero
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Nuria Baixeras
- Pathology Department, Bellvitge University Hospital, Hospitalet de Llobregat, Barcelona, Spain
| | - Anna Esteve-Codina
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.,Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Ernest Nadal
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Hospitalet de Llobregat, Barcelona, Spain.,Clinical Research in Solid Tumors (CReST) Group, OncoBell Program, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Elisabeth Brambilla
- Département d'Anatomie et Cytologie Pathologiques, Pôle de Biologie et Pathologie, CHU Grenoble Alpes, Grenoble and Université Grenoble Alpes, Grenoble, France
| | - Montse Sanchez-Cespedes
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
173
|
Jørgensen JT, Hersom M. Clinical and Regulatory Aspects of Companion Diagnostic Development in Oncology. Clin Pharmacol Ther 2018; 103:999-1008. [PMID: 29197081 DOI: 10.1002/cpt.955] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/16/2017] [Accepted: 11/27/2017] [Indexed: 12/21/2022]
Abstract
Nearly 20 years have passed since the US Food and Drug Administration (FDA) approved the first companion diagnostic and today this type of assay governs the use of 21 different anticancer drugs. The regulators deem these assays essential for the safe and effective use of a corresponding therapeutic product. The companion diagnostic assays are important both during the drug development process as well as essential treatment decision tools after the approval of the drugs.
Collapse
Affiliation(s)
| | - Maria Hersom
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
174
|
Katz SI, Hammer M, Bagley SJ, Aggarwal C, Bauml JM, Thompson JC, Nachiappan AC, Simone CB, Langer CJ. Radiologic Pseudoprogression during Anti-PD-1 Therapy for Advanced Non-Small Cell Lung Cancer. J Thorac Oncol 2018; 13:978-986. [PMID: 29738824 DOI: 10.1016/j.jtho.2018.04.010] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 04/01/2018] [Accepted: 04/06/2018] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Anti-programmed cell death protein 1 (PD-1) therapy can lead to unconventional tumor responses, including radiologic pseudoprogression. Here we have determined the real-world incidence of radiologic pseudoprogression in advanced NSCLC and compared radiologic response criteria for assessment of disease response. METHODS The electronic medical records of all patients with NSCLC who were receiving anti-PD-1 therapy at our institution over a 3-year period were retrospectively reviewed, and patients with clinically suspected radiologic pseudoprogression were identified. Patients without available follow-up imaging or clinical data were excluded. Imaging examinations were then analyzed to determine whether progression was confirmed on subsequent reimaging. Assessments of tumor response by the Response Evaluation Criteria in Solid Tumors (RECIST), version 1.1 (RECIST 1.1), the unidimensional immune-related response criteria (iRRC), and the iRECIST criteria for all patients were performed and compared. RESULTS A total of 228 consecutive patients began receiving anti-PD-1 therapy over a 3-year period. Of the 166 of these patients who were evaluable, most (80%) received nivolumab. Fifteen patients (9%) were clinically suspected of having radiologic pseudoprogression on account of tumor enlargement and/or development of new lesions on computed tomography images during the first 4 to 6 weeks of therapy, and they continued receiving anti-PD-1 therapy. Three of these patients (2%) demonstrated evidence of radiologic pseudoprogression at first reimaging. The iRRC and immune RECIST criteria were more accurate in classifying radiologic pseudoprogression as nonprogression; none of the three cases were deemed progression by the iRRC or immune RECIST, whereas all three cases were determined to be progression according to the Response Evaluation Criteria in Solid Tumors, version 1.1. CONCLUSIONS Radiologic pseudoprogression is a clinical challenge but an uncommon occurrence in patients with NSCLC who are receiving anti-PD-1 therapy.
Collapse
Affiliation(s)
- Sharyn I Katz
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania.
| | - Mark Hammer
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania; Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Stephen J Bagley
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Charu Aggarwal
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Joshua M Bauml
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Jeffrey C Thompson
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Arun C Nachiappan
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Charles B Simone
- Department of Radiation Oncology, University of Maryland Medical Center, Baltimore, Maryland
| | - Corey J Langer
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
175
|
Jaafar J, Fernandez E, Alwan H, Philippe J. Programmed cell death-1 and programmed cell death ligand-1 antibodies-induced dysthyroidism. Endocr Connect 2018; 7:R196-R211. [PMID: 29739808 PMCID: PMC5937198 DOI: 10.1530/ec-18-0079] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 04/09/2018] [Indexed: 12/28/2022]
Abstract
BACKGROUND Monoclonal antibodies blocking the programmed cell death-1 (PD-1) or its ligand (PD-L1) are a group of immune checkpoints inhibitors (ICIs) with proven antitumor efficacy. However, their use is complicated by immune-related adverse events (irAEs), including endocrine adverse events (eAEs). PURPOSE We review the incidence, time to onset and resolution rate of dysthyroidism induced by PD-1/PD-L1 Ab, and the clinical, biological and radiological findings. We aim to discuss the potential mechanisms of PD-1/PD-L1 Ab-induced dysthyroidism, and to propose a management algorithm. METHODS We performed a literature search of available clinical trials regarding PD-1/PD-L1 Ab in the PubMed database. We selected all English language clinical trials that included at least 100 patients. We also present selected case series or reports, retrospective studies and reviews related to this issue. FINDINGS In patients treated with PD-1 Ab, hypothyroidism occurred in 2-10.1% and hyperthyroidism occurred in 0.9-7.8%. When thyroiditis was reported separately, it occurred in 0.34-2.6%. Higher rates were reported when PD-1 Ab were associated with other ICI or chemotherapy. The median time to onset of hyperthyroidism and hypothyroidism after PD-1 Ab initiation was 23-45 days and 2-3.5 months, respectively. Regarding PD-L1 Ab, hypothyroidism occurred in 0-10% and hyperthyroidism in 0.5-2% of treated patients. The average time to onset of dysthyroidism after PD-L1 Ab was variable and ranged from 1 day after treatment initiation to 31 months. CONCLUSION Dysthyroidism occurs in up to 10% of patients treated with PD-1/PD-L1 Ab. Hypothyroidism and reversible destructive thyroiditis are the most frequent endocrine adverse events (eAE) in PD-1/PD-L1 treated patients. Immune and non-immune mechanisms are potentially involved, independently of the presence of thyroid antibodies.
Collapse
Affiliation(s)
- Jaafar Jaafar
- Division of Endocrinology and DiabetologyGeneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Eugenio Fernandez
- Department of OncologyGeneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Heba Alwan
- Division of Endocrinology and DiabetologyGeneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Jacques Philippe
- Division of Endocrinology and DiabetologyGeneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| |
Collapse
|
176
|
Affiliation(s)
- S Dacic
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, USA.
| |
Collapse
|
177
|
Cushman TR, Gomez D, Kumar R, Likacheva A, Chang JY, Cadena AP, Paris S, Welsh JW. Combining radiation plus immunotherapy to improve systemic immune response. J Thorac Dis 2018; 10:S468-S479. [PMID: 29593892 DOI: 10.21037/jtd.2018.01.130] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Over the past decade, the fields of oncology have made great strides in therapies. The development of new therapeutics and increased understanding of the role of the immune system in the development and treatment of cancer has led to increased collaboration between oncologic fields. Recent technologic advancements in radiation therapy (RT), including stereotactic beam radiation therapy (SBRT), have improved local control and offer an alternative to surgery for the control of oligometastatic disease. Immunotherapy has proven a promising therapeutic in the treatment of metastatic disease but treatment resistance remains a significant obstacle in the majority of patients. Together, radiation and immunotherapy offer potential to eliminate metastatic disease, reduce time to recurrence and improve overall survival. Major obstacles to these positive outcomes include high tumor burden, intratumoral heterogeneity, and the negative effects of tumor stroma, to name a few. Multimodality treatments are under heavy investigation. Promising data from clinical trials is emerging to highlight the value of RT in combination with immunotherapy. However, the mechanisms behind their synergistic effects remain to be fully elucidated. This review aims to highlight the existing literature and offers hypotheses to explain mechanisms behind the synergy of RT and immunotherapy.
Collapse
Affiliation(s)
- Taylor R Cushman
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel Gomez
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rachit Kumar
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Radiation Oncology, Banner MD Anderson Cancer Center, Phoenix, AZ, USA
| | - Anna Likacheva
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Radiation Oncology, Banner MD Anderson Cancer Center, Phoenix, AZ, USA
| | - Joe Y Chang
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alex P Cadena
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - James W Welsh
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
178
|
Hammer M, Bagley S, Aggarwal C, Bauml J, Nachiappan AC, Simone CB, Langer C, Katz SI. Thoracic Imaging of Non-Small Cell Lung Cancer Treated With Anti-programmed Death Receptor-1 Therapy. Curr Probl Diagn Radiol 2018; 48:142-147. [PMID: 29573843 DOI: 10.1067/j.cpradiol.2018.01.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 01/08/2018] [Indexed: 01/09/2023]
Abstract
PURPOSE Treatment with anti-programmed death receptor-1 (PD-1) therapeutics can lead to unconventional responses and side effect profiles due to their potentiating effects on the immune system. Here we evaluate the radiologic manifestations of anti-PD-1 therapy in the chest in patients with non-small cell lung cancer (NSCLC) receiving anti-PD-1 therapy. MATERIALS AND METHODS A retrospective review of real-world clinical practice was conducted of all the patients with NSCLC receiving anti-PD-1 therapy at our institution between 2013 and 2016. All patients without adequate clinical or radiologic follow-up data in the electronic medical records were excluded. Imaging examinations for all patients deemed by their thoracic oncologists to have radiologic pseudoprogression or therapy-associated pneumonitis were reviewed by experienced thoracic radiologists. RESULTS A total of 166 patients with NSCLC had available clinical and imaging data for retrospective review. Of these patients, 4 (2%) were considered to have radiologic pseudoprogression, 3 of which manifested as increased tumor size and 1 of which manifested with new lesions. A total of 5 patients (3%) were clinically deemed to have pneumonitis attributable to anti-PD-1 therapy, 4 of which had radiologic manifestations on computed tomography. CONCLUSION Radiologic pseudoprogression and drug-induced pneumonitis are uncommon but important manifestations of anti-PD-1 therapy on thoracic imaging.
Collapse
Affiliation(s)
- Mark Hammer
- Department of Radiology, Brigham and Women's Hospital, Boston, MA; Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Stephen Bagley
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Charu Aggarwal
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Joshua Bauml
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Arun C Nachiappan
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Charles B Simone
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore, MD
| | - Corey Langer
- Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Sharyn I Katz
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA.
| |
Collapse
|
179
|
Zou T, Awad MM. More valuable than platinum: first-line pembrolizumab in advanced stage non-small-cell lung cancer. Ann Oncol 2018; 28:685-687. [PMID: 28327984 DOI: 10.1093/annonc/mdx083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Indexed: 01/09/2023] Open
Affiliation(s)
- T Zou
- Dana Farber Cancer Institute, Medical Oncology
| | - M M Awad
- Dana Farber Cancer Institute, Medical Oncology
| |
Collapse
|
180
|
Zheng Y, Narwal R, Jin C, Baverel PG, Jin X, Gupta A, Ben Y, Wang B, Mukhopadhyay P, Higgs BW, Roskos L. Population Modeling of Tumor Kinetics and Overall Survival to Identify Prognostic and Predictive Biomarkers of Efficacy for Durvalumab in Patients With Urothelial Carcinoma. Clin Pharmacol Ther 2018; 103:643-652. [PMID: 29243222 PMCID: PMC5873369 DOI: 10.1002/cpt.986] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/11/2017] [Accepted: 12/12/2017] [Indexed: 12/13/2022]
Abstract
Durvalumab is an anti‐PD‐L1 monoclonal antibody approved for patients with locally advanced or metastatic urothelial carcinoma (UC) that has progressed after platinum‐containing chemotherapy. A population tumor kinetic model, coupled with dropout and survival models, was developed to describe longitudinal tumor size data and predict overall survival in UC patients treated with durvalumab (NCT01693562) and to identify prognostic and predictive biomarkers of clinical outcomes. Model‐based covariate analysis identified liver metastasis as the most influential factor for tumor growth and immune‐cell PD‐L1 expression and baseline tumor burden as predictive factors for tumor killing. Tumor or immune‐cell PD‐L1 expression, liver metastasis, baseline hemoglobin, and albumin levels were identified as significant covariates for overall survival. These model simulations provided further insights into the impact of PD‐L1 cutoff values on treatment outcomes. The modeling framework can be a useful tool to guide patient selection and enrichment strategies for immunotherapies across various cancer indications.
Collapse
Affiliation(s)
| | | | - ChaoYu Jin
- MedImmune, Mountain View, California, USA
| | | | | | | | | | - Bing Wang
- MedImmune, Mountain View, California, USA
| | | | | | | |
Collapse
|
181
|
Liu J, Zhang C, Hu J, Tian Q, Wang X, Gu H, Zhang S, Zhao D, Fan R. Effectiveness of anti-PD-1/PD-L1 antibodies in urothelial carcinoma patients with different PD-L1 expression levels: a meta-analysis. Oncotarget 2018; 9:12400-12407. [PMID: 29552320 PMCID: PMC5844756 DOI: 10.18632/oncotarget.24249] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 12/05/2017] [Indexed: 02/01/2023] Open
Abstract
Background Urothelial carcinoma ranks the ninth among malignant cancers. We conducted this study to identify which patients could benefit more from the treatment of programmed death-1 (PD–1)/programmed death-ligand1 (PD–L1) inhibitors. Materials and Methods We performed literature searches, combined data from qualified literature and performed comparative analyses on the effectiveness of anti-PD–1/PD–L1 antibodies in patients with different PD–L1 expression levels. Results We divided patients into three groups according to the percentages of PD–L1-positive cells, namely the low- PD-L1 (PD-L1 < 1%), the medium-PD-L1 (PD-L1 ≥ 1 and < 5%) and the high–PD–L1 (PD-L1 ≥ 5%) groups. We found that the high-PD-L1 group responded significantly better than other groups (P = 0.0003, ORs = 0.45, 95%CI: 0.29-071; P = 0.0009, ORs = 0.43, 95%CI: 0.25-0.73, for low-PD-L1 and medium-PD-L1 groups, respectively), while the latter two groups responded similarly (P = 0.90, ORs = 1.06, 95%CI: 0.62-1.83) to both PD–1 and PD–L1 inhibitors. Furthermore, we found that the medium-PD–L1 and high-PD–L1 groups responded similarly to PD-1/ PD-L1 inhibitors (P = 0.65, ORs = 1.11, 95%CI: 0.69–1.77), while the low-PD–L1 group responded better to PD-1 inhibitors than PD-L1 inhibitors (P = 0.046, ORs = 1.92, 95%CI: 0.98–3.89). Conclusions Our results suggest that PD–L1 positive patients should be defined as those with ≥ 5% or greaterPD-L1-positive cells. PD-1 antibodies performed better only in the low-group patients, likely because they could block the interactions of PD–1 with both PD–L1 and PD–L2.
Collapse
Affiliation(s)
- Junqi Liu
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, 450000 Zhengzhou, Henan, China
| | - Chuanfeng Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, 450000 Zhengzhou, Henan, China
| | - Jiegang Hu
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, 450000 Zhengzhou, Henan, China
| | - Qing Tian
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, 450000 Zhengzhou, Henan, China
| | - Xin Wang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, 450000 Zhengzhou, Henan, China
| | - Hao Gu
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, 450000 Zhengzhou, Henan, China
| | - Song Zhang
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, 450000 Zhengzhou, Henan, China
| | - Di Zhao
- Endocrinology Department, The First Affiliated Hospital of Zhengzhou University, 450000 Zhengzhou, Henan, China
| | - Ruitai Fan
- Department of Radiotherapy, The First Affiliated Hospital of Zhengzhou University, 450000 Zhengzhou, Henan, China
| |
Collapse
|
182
|
Annunziata CM, Kohn EC. Clinical trials in gynecologic oncology: Past, present, and future. Gynecol Oncol 2017; 148:393-402. [PMID: 29212614 DOI: 10.1016/j.ygyno.2017.11.026] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/17/2017] [Accepted: 11/20/2017] [Indexed: 12/18/2022]
Abstract
The Gynecologic Oncology Group has historically performed ground-breaking, practice-changing clinical trials in women's cancers. The current standard of care for initial treatment of ovarian, endometrial, cervical, and trophoblastic cancers was determined by clinical trials completed within this cooperative group structure. For example, trial GOG-0111 set the standard for combining platinum and taxane chemotherapy in ovarian cancer, and more recently GOG-0240 provided evidence for adding bevacizumab to chemotherapy for women with advanced cervical cancer. The landscape of clinical trial design has markedly changed in recent decades, with a clear emphasis on streamlining drug development towards specific patient populations and indications for investigational agents. Translational science in gynecologic cancers can set the stage for rapid and efficient introduction of new therapies for our patients. The gynecologic oncology community of researchers and clinicians is well positioned to enter into the new era of drug development, with breakthrough discoveries increasing each year. It is clear that we must incorporate smarter clinical trial design to get the right drugs to the right patients expeditiously, so we can continue to improve outcome for women with gynecologic cancers.
Collapse
Affiliation(s)
- Christina M Annunziata
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States.
| | - Elise C Kohn
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, United States
| |
Collapse
|
183
|
Assi HI, Kamphorst AO, Moukalled NM, Ramalingam SS. Immune checkpoint inhibitors in advanced non-small cell lung cancer. Cancer 2017; 124:248-261. [PMID: 29211297 DOI: 10.1002/cncr.31105] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 09/22/2017] [Accepted: 10/03/2017] [Indexed: 12/17/2022]
Abstract
The emergence of immune checkpoint inhibitors for the treatment of cancer has led to major changes to the therapeutic landscape of lung cancer. Improvements in overall survival relative to standard chemotherapy have been observed in the first-line and second-line therapy settings for patients with advanced non-small cell lung cancer (NSCLC) who are treated with immune checkpoint inhibitors. Consequently, every patient with advanced-stage NSCLC is now a candidate for immune checkpoint inhibitor therapy. However, it is clear that the benefit from therapy is not universal, and identification of biomarkers to select therapy has assumed importance. In addition to programmed cell death receptor ligand 1 expression, both tissue-based and blood-based markers are under evaluation to select patients. In an era of increasing costs of care and potential for toxicities related to immune checkpoint inhibition, proper patient selection is critical to the optimal use of this new class of agents. In addition, development of novel combination approaches has also emerged as an important way to improve the efficacy of immune checkpoint inhibition. Studies in earlier stages of NSCLC are already underway with the hope of improving the cure rate. In this article, the authors review the current landscape of immune checkpoint inhibitors in the treatment of advanced NSCLC. Cancer 2018;124:248-61. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- Hazem I Assi
- Department of Internal Medicine, Naef K. Bassile Cancer Institute, American University of Beirut, Beirut, Lebanon
| | - Alice O Kamphorst
- Department of Microbiology and Immunology, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia
| | - Nour M Moukalled
- Department of Internal Medicine, Naef K. Bassile Cancer Institute, American University of Beirut, Beirut, Lebanon
| | - Suresh S Ramalingam
- Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
184
|
Reck M. Pembrolizumab as first-line therapy for metastatic non-small-cell lung cancer. Immunotherapy 2017; 10:93-105. [PMID: 29145737 DOI: 10.2217/imt-2017-0121] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
This review describes trials evaluating the monoclonal antibody pembrolizumab (an immunotherapy that blocks the interaction between programmed death-1 and programmed death-ligand 1 and 2 [PD-L1/PD-L2]) as first-line therapy for advanced non-small-cell lung cancer (NSCLC). In the Phase III KEYNOTE-024 study, pembrolizumab monotherapy significantly improved progression-free survival (primary end point) and overall survival, and was associated with fewer adverse events compared with platinum-based chemotherapy in patients with NSCLC with PD-L1 expression on ≥50% of tumor cells. In cohort G of the Phase I/II KEYNOTE-021 study, pembrolizumab plus pemetrexed and carboplatin significantly improved objective response rate (primary end point) and progression-free survival versus pemetrexed and carboplatin alone, and had manageable toxicity in patients with nonsquamous NSCLC. These results have changed first-line management of advanced NSCLC.
Collapse
Affiliation(s)
- Martin Reck
- Lung Clinic Grosshansdorf, Airway Research Center North (ARCN), German Center of Lung Research (DZL), Grosshansdorf, Germany
| |
Collapse
|
185
|
Hofman P. PD-L1 immunohistochemistry for non-small cell lung carcinoma: which strategy should be adopted? Expert Rev Mol Diagn 2017; 17:1097-1108. [PMID: 29069958 DOI: 10.1080/14737159.2017.1398083] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION PD-L1 detection with immunohistochemistry (IHC) is the only predictive biomarker available to date for PD-L1/PD1 immunotherapy in thoracic oncology. While many studies have been published on this biomarker, they raise a number of questions concerning mainly, (i) the type of antibody for use and its condition of utilization, (ii) the threshold to be used, (iii) the message and information to communicate to the thoracic oncologist and, (iv) the adoption of this methodology as part of the daily practices of a pathology laboratory. Areas covered: This review provides an update on the use of the different PD-L1 antibodies for IHC in the context of metastatic non-small cell lung cancer (NSCLC) and discusses their use as companion or complementary diagnostic tests. The limits of PD-L1 IHC as a predictive test, the precautions to be adopted as well as some perspectives will then be considered. Expert commentary: IHC for PD-L1 can be considered as a theranostic test, which implies providing an extremely reliable result that avoids any false positive and negative results. PD-L1 IHC requires considerable expertise and specific training of pathologists. PD-L1 IHC can be a companion or complementary diagnostic test depending on the clone employed, the molecular therapy prescribed and the indication of use.
Collapse
Affiliation(s)
- Paul Hofman
- a Laboratoire de Pathologie Clinique et Expérimentale, Hôpital Pasteur , Centre Hospitalo-Universitaire de Nice, Université de Nice Sophia Antipolis , Nice cedex 01 , France.,b Biobanque Hospitalière (BB-0033-00025) , Hôpital Pasteur , Nice , France.,c FHU OncoAge , Hôpital Pasteur, Université Nice Côte d'Azur , Nice , France
| |
Collapse
|
186
|
Li D, Wang W. Booming cancer immunotherapy fighting tumors. SCIENCE CHINA-LIFE SCIENCES 2017; 60:1445-1449. [DOI: 10.1007/s11427-017-9208-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 10/16/2017] [Indexed: 12/30/2022]
|
187
|
Wang PF, Chen Y, Song SY, Wang TJ, Ji WJ, Li SW, Liu N, Yan CX. Immune-Related Adverse Events Associated with Anti-PD-1/PD-L1 Treatment for Malignancies: A Meta-Analysis. Front Pharmacol 2017; 8:730. [PMID: 29093678 PMCID: PMC5651530 DOI: 10.3389/fphar.2017.00730] [Citation(s) in RCA: 343] [Impact Index Per Article: 42.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 09/28/2017] [Indexed: 12/12/2022] Open
Abstract
Background: Treatment of cancers with programmed cell death protein 1 (PD-1) pathway inhibitors can lead to immune-related adverse events (irAEs), which could be serious and even fetal. Therefore, clinicians should be aware of the characteristics of irAEs associated with the use of such drugs. Methods: The MEDLINE, EMBASE, and Cochrane databases were searched to find potential studies using the following strategies: anti-PD-1/PD-L1 treatment; irAEs; and cancer. R© package Meta was used to pool incidence. Results: Forty-six studies representing 12,808 oncologic patients treated with anti-PD-1/PD-L1 agents were included in the meta-analysis. The anti-PD-1/PD-L1 agents included nivolumab, pembrolizumab, atezolizumab, durvalumab, avelumab, and BMS-936559. The tumor types were melanomas, Hodgkin lymphomas, urothelial carcinomas, breast cancers, non-small cell lung cancers, renal cell carcinomas (RCC), colorectal cancers, and others. We described irAEs according to organ systems, namely, the skin (pruritus, rash, maculopapular rash, vitiligo, and dermatitis), endocrine system (hypothyroidism, hyperthyroidism, hypophysitis, thyroiditis, and adrenal insufficiency), digestive system (colitis, diarrhea, pancreatitis, and increased AST/ALT/bilirubin), respiratory system (pneumonitis, lung infiltration, and interstitial lung disease), and urinary system (increased creatinine, nephritis, and renal failure). In patients treated with the PD-1 signaling inhibitors, the overall incidence of irAEs was 26.82% (95% CI, 21.73-32.61; I2, 92.80) in any grade and 6.10% (95% CI, 4.85-7.64; I2, 52.00) in severe grade, respectively. The development of irAEs was unrelated to the dose of anti-PD-1/PD-L1 agents. The incidence of particular irAEs varied when different cancers were treated with different drugs. The incidence of death due to irAEs was around 0.17%. Conclusion: The occurrence of irAEs was organ-specific and related to drug and tumor types.
Collapse
Affiliation(s)
- Peng-Fei Wang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Yang Chen
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China.,State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Si-Ying Song
- Department of Clinical Medicine, Capital Medical University, Beijing, China
| | - Ting-Jian Wang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Wen-Jun Ji
- Key Laboratory, Department of Neurosurgery, The Second Hospital of Yulin, Xi'an Jiaotong University, Xi'an, China
| | - Shou-Wei Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Ning Liu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| | - Chang-Xiang Yan
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
188
|
PD-1/PD-L1 checkpoint blockades in non-small cell lung cancer: New development and challenges. Cancer Lett 2017; 405:29-37. [DOI: 10.1016/j.canlet.2017.06.033] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/21/2017] [Accepted: 06/28/2017] [Indexed: 11/18/2022]
|
189
|
Zarogoulidis P, Papadopoulos V, Maragouli E, Papatsibas G, Huang H. Checkpoint inhibitors in metastatic epidermal growth factor receptor-mutated non-small cell lung cancer patients: where we treating the wrong cancer? J Thorac Dis 2017; 9:2771-2773. [PMID: 29221236 DOI: 10.21037/jtd.2017.08.24] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Paul Zarogoulidis
- Pulmonary Department-Oncology Unit, "Theageneio" Anticancer Hospital, Thessaloniki, Greece
| | | | - Elena Maragouli
- Oncology Department, University of Thessali, Larissa, Greece
| | | | - Haidong Huang
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| |
Collapse
|
190
|
Vanpouille-Box C, Lhuillier C, Bezu L, Aranda F, Yamazaki T, Kepp O, Fucikova J, Spisek R, Demaria S, Formenti SC, Zitvogel L, Kroemer G, Galluzzi L. Trial watch: Immune checkpoint blockers for cancer therapy. Oncoimmunology 2017; 6:e1373237. [PMID: 29147629 DOI: 10.1080/2162402x.2017.1373237] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 08/25/2017] [Indexed: 02/08/2023] Open
Abstract
Immune checkpoint blockers (ICBs) are literally revolutionizing the clinical management of an ever more diversified panel of oncological indications. Although considerable attention persists around the inhibition of cytotoxic T lymphocyte-associated protein 4 (CTLA4) and programmed cell death 1 (PDCD1, best known as PD-1) signaling, several other co-inhibitory T-cell receptors are being evaluated as potential targets for the development of novel ICBs. Moreover, substantial efforts are being devoted to the identification of biomarkers that reliably predict the likelihood of each patient to obtain clinical benefits from ICBs in the absence of severe toxicity. Tailoring the delivery of specific ICBs or combinations thereof to selected patient populations in the context of precision medicine programs constitutes indeed a major objective of the future of ICB-based immunotherapy. Here, we discuss recent preclinical and clinical advances on the development of ICBs for oncological indications.
Collapse
Affiliation(s)
| | - Claire Lhuillier
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Lucillia Bezu
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | - Fernando Aranda
- Immunoreceptors of the Innate and Adaptive System Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Oliver Kepp
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
| | - Jitka Fucikova
- Sotio a.c., Prague, Czech Republic.,Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Radek Spisek
- Sotio a.c., Prague, Czech Republic.,Dept. of Immunology, 2nd Faculty of Medicine and University Hospital Motol, Charles University, Prague, Czech Republic
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,INSERM, U1015, Villejuif, France.,Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France.,Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, Paris, France.,Université Pierre et Marie Curie/Paris VI, Paris, France.,Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France.,INSERM, U1138, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden.,Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP, Paris, France
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Université Paris Descartes/Paris V, Paris, France.,Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
191
|
Abstract
Introduction Immunotherapy in the form of immune checkpoint inhibitors has changed the landscape of cancer treatment. Newer monoclonal antibodies are coming up and are being tested in various cancers during different stages of treatment. With the increasing use of immune checkpoint inhibitors in the management of various types of cancers, the question is raised as to what next can be offered to a patient who has progressed on this newer treatment. Does Sequence matter? There have been reports of improved responses to chemotherapy after immunotherapy in the form of vaccines. Here we present a case series of 6 patients who progressed on immunotherapy with immune checkpoint inhibitors after initial modality of treatment (chemotherapy/radiation), subsequently received chemotherapy with excellent response. Methods We have a cohort of six patients who had disease progression on second line Immunotherapy for solid or hematological malignancies and had ECOG < 2. All these patients received third line salvage chemotherapy. Three patients had metastatic head and neck cancer, 2 had non-small cell lung cancer (NSCLC), and one had T -cell rich B- cell lymphoma. Prior review and approval were obtained from our institutional review board. Results All patients had an excellent response to chemotherapy in third line setting, after immune checkpoint inhibitors and most of them achieved a complete response. Conclusion Targeting cancer with chemotherapy after failure of immunotherapy is a valid option and can lead to better response rates and PFS which may lead to OS. This effect may be secondary to immunotherapy removing the inhibition exerted by tumor cells or other immune cells initially followed by cytotoxic chemotherapy mediated killing of tumor cells.
Collapse
|
192
|
Cummings AL, Garon EB. The ascent of immune checkpoint inhibitors: is the understudy ready for a leading role? Cancer Biol Med 2017; 14:341-347. [PMID: 29372100 PMCID: PMC5785166 DOI: 10.20892/j.issn.2095-3941.2017.0090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Affiliation(s)
- Amy L Cummings
- Department of Hematology and Oncology, David Geffen School of Medicine at the University of California, Los Angeles 90095, USA
| | - Edward B Garon
- Department of Hematology and Oncology, David Geffen School of Medicine at the University of California, Los Angeles 90095, USA
| |
Collapse
|