101
|
Comparison of Different Methods for Defining Hyperprogressive Disease in NSCLC. JTO Clin Res Rep 2021; 2:100115. [PMID: 34589976 PMCID: PMC8474364 DOI: 10.1016/j.jtocrr.2020.100115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/06/2020] [Accepted: 10/20/2020] [Indexed: 01/28/2023] Open
Abstract
Introduction Hyperprogressive disease (HPD) as a consequence of immune checkpoint inhibitors in NSCLC has been reported in multiple studies. However, inconsistent results in incidence and survival outcomes within studies, together with different assessment methods, have led to increasing controversy regarding the concept of HPD. Methods Consecutive patients treated with nivolumab (N = 42) or docetaxel (N = 37) were evaluated. HPD was quantified by applying three different methods (tumor growth rate [TGR], tumor growth kinetics [TGK], and Response Evaluation Criteria in Solid Tumors version 1.1 [RECIST 1.1]). HPD rates were compared between and within both cohorts using the different methods. Results Using TGR, TGK, and RECIST 1.1, we identified seven (16.7%), seven (16.7%), and six (14.3%) patients with HPD in the nivolumab cohort and three (8.1%), four (10.8%), and five (13.6%) in the docetaxel cohort, respectively. We observed a higher concordance between TGR and TGK (90.1%) compared with RECIST 1.1 (31.3% and 37.5% with TGR and TGK, respectively). We found no significant differences in the overall survival between patients with progressive disease and HPD in either cohort. Conclusions TGR and TGK revealed high concordance rates for identifying patients with HPD in NSCLC. The incidence of HPD was numerically higher in patients treated with immune checkpoint inhibitors. Standardization of methods for measuring HPD and its exploration in larger studies are needed to establish its clinical meaning in NSCLC.
Collapse
|
102
|
Chemotherapy Should Be Combined With Checkpoint Inhibitors in the Treatment of Patients With Stage IV EGFR-Mutant NSCLC Whose Disease Has Progressed on All Available Tyrosine Kinase Inhibitors. J Thorac Oncol 2021; 16:1622-1626. [PMID: 34561035 DOI: 10.1016/j.jtho.2021.07.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/09/2021] [Accepted: 07/15/2021] [Indexed: 02/04/2023]
|
103
|
An HJ, Chon HJ, Kim C. Peripheral Blood-Based Biomarkers for Immune Checkpoint Inhibitors. Int J Mol Sci 2021; 22:9414. [PMID: 34502325 PMCID: PMC8430528 DOI: 10.3390/ijms22179414] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 02/08/2023] Open
Abstract
As cancer immunotherapy using immune checkpoint inhibitors (ICIs) is rapidly evolving in clinical practice, it is necessary to identify biomarkers that will allow the selection of cancer patients who will benefit most or least from ICIs and to longitudinally monitor patients' immune responses during treatment. Various peripheral blood-based immune biomarkers are being identified with recent advances in high-throughput multiplexed analytical technologies. The identification of these biomarkers, which can be easily detected in blood samples using non-invasive and repeatable methods, will contribute to overcoming the limitations of previously used tissue-based biomarkers. Here, we discuss the potential of circulating immune cells, soluble immune and inflammatory molecules, circulating tumor cells and DNA, exosomes, and the blood-based tumor mutational burden, as biomarkers for the prediction of immune responses and clinical benefit from ICI treatment in patients with advanced cancer.
Collapse
Affiliation(s)
- Ho Jung An
- Department of Medical Oncology, St. Vincent’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea;
| | - Hong Jae Chon
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam 13496, Korea
| | - Chan Kim
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam 13496, Korea
| |
Collapse
|
104
|
Martinez-Usatorre A, Kadioglu E, Boivin G, Cianciaruso C, Guichard A, Torchia B, Zangger N, Nassiri S, Keklikoglou I, Schmittnaegel M, Ries CH, Meylan E, De Palma M. Overcoming microenvironmental resistance to PD-1 blockade in genetically engineered lung cancer models. Sci Transl Med 2021; 13:13/606/eabd1616. [PMID: 34380768 DOI: 10.1126/scitranslmed.abd1616] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 03/23/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022]
Abstract
Immune checkpoint blockade (ICB) with PD-1 or PD-L1 antibodies has been approved for the treatment of non-small cell lung cancer (NSCLC). However, only a minority of patients respond, and sustained remissions are rare. Both chemotherapy and antiangiogenic drugs may improve the efficacy of ICB in mouse tumor models and patients with cancer. Here, we used genetically engineered mouse models of Kras G12D/+;p53 -/- NSCLC, including a mismatch repair-deficient variant (Kras G12D/+;p53 -/-;Msh2 -/-) with higher mutational burden, and longitudinal imaging to study tumor response and resistance to combinations of ICB, antiangiogenic therapy, and chemotherapy. Antiangiogenic blockade of vascular endothelial growth factor A and angiopoietin-2 markedly slowed progression of autochthonous lung tumors, but contrary to findings in other cancer types, addition of a PD-1 or PD-L1 antibody was not beneficial and even accelerated progression of a fraction of the tumors. We found that antiangiogenic treatment facilitated tumor infiltration by PD-1+ regulatory T cells (Tregs), which were more efficiently targeted by the PD-1 antibody than CD8+ T cells. Both tumor-associated macrophages (TAMs) of monocyte origin, which are colony-stimulating factor 1 receptor (CSF1R) dependent, and TAMs of alveolar origin, which are sensitive to cisplatin, contributed to establish a transforming growth factor-β-rich tumor microenvironment that supported PD-1+ Tregs Dual TAM targeting with a combination of a CSF1R inhibitor and cisplatin abated Tregs, redirected the PD-1 antibody to CD8+ T cells, and improved the efficacy of antiangiogenic immunotherapy, achieving regression of most tumors.
Collapse
Affiliation(s)
- Amaia Martinez-Usatorre
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland.,Agora Cancer Research Center, 1011 Lausanne, Switzerland
| | - Ece Kadioglu
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Gael Boivin
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland.,Agora Cancer Research Center, 1011 Lausanne, Switzerland
| | - Chiara Cianciaruso
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland.,Agora Cancer Research Center, 1011 Lausanne, Switzerland
| | - Alan Guichard
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland.,Agora Cancer Research Center, 1011 Lausanne, Switzerland
| | - Bruno Torchia
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland.,Agora Cancer Research Center, 1011 Lausanne, Switzerland
| | - Nadine Zangger
- Bioinformatics Core Facility (BCF), SIB Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Sina Nassiri
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland.,Agora Cancer Research Center, 1011 Lausanne, Switzerland.,Bioinformatics Core Facility (BCF), SIB Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Ioanna Keklikoglou
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Martina Schmittnaegel
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland.,Roche Innovation Center Munich, Oncology Discovery, Pharma Research and Early Development, 82377 Penzberg, Germany
| | - Carola H Ries
- Roche Innovation Center Munich, Oncology Discovery, Pharma Research and Early Development, 82377 Penzberg, Germany
| | - Etienne Meylan
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Michele De Palma
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology in Lausanne (EPFL), 1015 Lausanne, Switzerland. .,Agora Cancer Research Center, 1011 Lausanne, Switzerland
| |
Collapse
|
105
|
Wang Y, Cuggia A, Pacis A, Boileau JC, Marcus VA, Gao ZH, Chong G, Foulkes WD, Zogopoulos G. Pancreatic Cancer Progression in a Patient With Lynch Syndrome Receiving Immunotherapy: A Cautionary Tale. J Natl Compr Canc Netw 2021; 19:883-887. [PMID: 34416708 DOI: 10.6004/jnccn.2021.7049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/21/2021] [Indexed: 11/17/2022]
Abstract
Pancreatic ductal adenocarcinomas (PDACs) with DNA mismatch repair deficiency (MMRd) respond preferentially to immune checkpoint inhibitors (ICIs). However, a subset of MMRd PDACs does not respond to these agents. This report describes a patient with PDAC who experienced rapid disease progression suggestive of hyperprogressive disease. The case involved a 63-year-old man carrying a pathogenic germline PMS2 mutation who developed metastatic PDAC. His tumor showed isolated loss of PMS2 expression by immunohistochemistry (IHC). He was treated with pembrolizumab, but his disease rapidly progressed. Whole-genome and transcriptome sequencing of a liver metastasis biopsy, acquired at disease progression, showed a retained wild-type PMS2 allele and hallmarks of microsatellite stability, including low tumor mutational burden and low MSIsensor score. PCR-based microsatellite instability (MSI) testing of the treatment-naïve tumor showed microsatellite stability. The ICI-treated tumor had a lower density of CD8+ T-cell infiltration than the treatment-naïve tumor, which is contrary to the expected evolution with ICI responsiveness. Through this case and a review of the literature, we highlight the low penetrance of PMS2 germline mutations in PDAC and discuss pitfalls in ascertaining MMRd and MSI based on IHC testing alone. An orthogonal confirmatory assay is warranted in the presence of uncommon immunophenotypes, such as isolated PMS2 loss, to optimize selection of patients with PDAC for immunotherapy.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Surgery, McGill University.,Research Institute of the McGill University Health Centre.,The Rosalind and Morris Goodman Cancer Research Centre, McGill University
| | - Adeline Cuggia
- Research Institute of the McGill University Health Centre
| | - Alain Pacis
- The Rosalind and Morris Goodman Cancer Research Centre, McGill University.,Canadian Centre for Computational Genomics, McGill University and Genome Quebec Innovation Center
| | | | | | - Zu-Hua Gao
- Research Institute of the McGill University Health Centre.,Department of Pathology, McGill University
| | - George Chong
- Molecular Diagnostics Laboratory, Sir Mortimer B. Davis-Jewish General Hospital; and
| | - William D Foulkes
- Research Institute of the McGill University Health Centre.,Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - George Zogopoulos
- Department of Surgery, McGill University.,Research Institute of the McGill University Health Centre.,The Rosalind and Morris Goodman Cancer Research Centre, McGill University.,Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
106
|
Combining Cancer Vaccines with Immunotherapy: Establishing a New Immunological Approach. Int J Mol Sci 2021; 22:ijms22158035. [PMID: 34360800 PMCID: PMC8348347 DOI: 10.3390/ijms22158035] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
Therapeutic cancer vaccines have become increasingly qualified for use in personalized cancer immunotherapy. A deeper understanding of tumor immunology and novel antigen delivery technologies has assisted in optimizing vaccine design. Therapeutic cancer vaccines aim to establish long-lasting immunological memory against tumor cells, thereby leading to effective tumor regression and minimizing non-specific or adverse events. However, due to several resistance mechanisms, significant challenges remain to be solved in order to achieve these goals. In this review, we describe our current understanding with respect to the use of the antigen repertoire in vaccine platform development. We also summarize various intrinsic and extrinsic resistance mechanisms behind the failure of cancer vaccine development in the past. Finally, we suggest a strategy that combines immune checkpoint inhibitors to enhance the efficacy of cancer vaccines.
Collapse
|
107
|
Moeller M, Adner J, Schuette W, Krueger M. [30- and 90-day Lethality in Patients with Stage IV Lung Cancer Depending on the Primary Therapy]. Pneumologie 2021; 75:950-959. [PMID: 34298565 DOI: 10.1055/a-1534-1339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Early lethality after initiation of therapy in patients with stage IV lung cancer has rarely been the focus of scientific studies yet. The little time remaining between diagnosis, start of therapy and onset of death, as well as any influencing factors, are of special interest for both, patients and physician. Accordingly, the aim of this work was to analyze the 30- and 90-day morbidity after initiation of systemic therapy and to determine possible factors influencing early lethality. For this purpose, the data of 225 patients with stage IV lung cancer and treatment at the Martha-Maria Halle-Dölau Lung Cancer Center between 01/01/2017 and 05/18/2020 were retrospectively analyzed. Forms of therapy and patient characteristics were analyzed with a frequency distribution and the probability of survival was estimated using the Kaplan-Meier method. The analysis of the early morbidity of all tumor-specifically treated patients showed a morbidity of 8.5 % at day 30 after the start of therapy and a rate of 23.5 % after 90 days. In a direct comparison of the different therapy groups, the patients receiving mono-checkpointinhibition had higher lethality (16.6 % after 30 days and 44.3 % after 90 days). In contrast, the morbidity of patients in the other therapy groups remained below 10 % after 30 days and below 23.3 % after 90 days. A poor general condition, an advanced tumor disease, polymetastasis and a positive history of smoking could be determined as predictors for higher early lethality. In contrast, there was no relevant difference in morbidity between the different tumor entities, gender, PD-L1 and mutation status. With this analysis, very high early lethality, comparable to other studies, could be detected in patients with lung cancer. Relevant differences between the forms of therapy illustrate the importance of individual patient selection for the respective therapy options and the rapid decision to initiate therapy.
Collapse
Affiliation(s)
- Miriam Moeller
- Klinik für Innere Medizin II, Krankenhaus Martha-Maria Halle-Dölau
| | - Juliane Adner
- Klinik für Innere Medizin II, Krankenhaus Martha-Maria Halle-Dölau
| | | | - Marcus Krueger
- Klinik für Thorxchirurgie, Krankenhaus Martha-Maria Halle-Dölau
| |
Collapse
|
108
|
Wang S, Xie K, Liu T. Cancer Immunotherapies: From Efficacy to Resistance Mechanisms - Not Only Checkpoint Matters. Front Immunol 2021; 12:690112. [PMID: 34367148 PMCID: PMC8335396 DOI: 10.3389/fimmu.2021.690112] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/05/2021] [Indexed: 01/05/2023] Open
Abstract
The immunotherapeutic treatment of various cancers with an increasing number of immune checkpoint inhibitors (ICIs) has profoundly improved the clinical management of advanced diseases. However, just a fraction of patients clinically responds to and benefits from the mentioned therapies; a large proportion of patients do not respond or quickly become resistant, and hyper- and pseudoprogression occur in certain patient populations. Furthermore, no effective predictive factors have been clearly screened or defined. In this review, we discuss factors underlying the elucidation of potential immunotherapeutic resistance mechanisms and the identification of predictive factors for immunotherapeutic responses. Considering the heterogeneity of tumours and the complex immune microenvironment (composition of various immune cell subtypes, disease processes, and lines of treatment), checkpoint expression levels may not be the only factors underlying immunotherapy difficulty and resistance. Researchers should consider the tumour microenvironment (TME) landscape in greater depth from the aspect of not only immune cells but also the tumour histology, molecular subtype, clonal heterogeneity and evolution as well as micro-changes in the fine structural features of the tumour area, such as myeloid cell polarization, fibroblast clusters and tertiary lymphoid structure formation. A comprehensive analysis of the immune and molecular profiles of tumour lesions is needed to determine the potential predictive value of the immune landscape on immunotherapeutic responses, and precision medicine has become more important.
Collapse
Affiliation(s)
- Shuyue Wang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, China
| | - Kun Xie
- German Cancer Research Center (DKFZ), Heidelberg University, Heidelberg, Germany
| | - Tengfei Liu
- Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
109
|
Kang BW, Chau I. Current status and future potential of predictive biomarkers for immune checkpoint inhibitors in gastric cancer. ESMO Open 2021; 5:S2059-7029(20)32652-1. [PMID: 32817133 PMCID: PMC7440716 DOI: 10.1136/esmoopen-2020-000791] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/17/2020] [Accepted: 05/25/2020] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy is revolutionising cancer treatment and has already emerged as standard treatment for patients with recurrent or metastatic gastric cancer (GC). Recent research has been focused on identifying robust predictive biomarkers for GC treated with immune checkpoint inhibitors (ICIs). The expression of programmed cell death protein-ligand-1 (PD-L1) is considered a manifestation of immune response evasion, and several studies have already reported the potential of PD-L1 expression as a predictive parameter for various human malignancies. Meanwhile, based on comprehensive molecular characterisation of GC, testing for Epstein-Barr virus and microsatellite instability is a potential predictive biomarker. Culminating evidence suggests that novel biomarkers, such as the tumour mutational burden and gene expression signature, could indicate the success of treatment with ICIs. However, the exact roles of these biomarkers in GC treated with ICIs remain unclear. Therefore, this study reviews recent scientific data on current and emerging biomarkers for ICIs in GC, which have potential to improve treatment outcomes.
Collapse
Affiliation(s)
- Byung Woog Kang
- Department of Oncology/Hematology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ian Chau
- Department of Medicine, Royal Marsden Hospital, London and Surrey, UK
| |
Collapse
|
110
|
Shen P, Han L, Ba X, Qin K, Tu S. Hyperprogressive Disease in Cancers Treated With Immune Checkpoint Inhibitors. Front Pharmacol 2021; 12:678409. [PMID: 34290608 PMCID: PMC8287409 DOI: 10.3389/fphar.2021.678409] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/22/2021] [Indexed: 12/24/2022] Open
Abstract
Immunotherapy, which takes advantage of the immune system to eliminate cancer cells, has been widely studied and applied in oncology. Immune checkpoint inhibitors (ICIs) prevent the immune system from being turned off before cancer cells are eliminated. They have proven to be among the most promising and effective immunotherapies, with significant survival benefits and durable responses in diverse tumor types. However, an increasing number of retrospective studies have found that some patients treated with ICIs experience unusual responses, including accelerated proliferation of tumor cells and rapid progression of the disease, with poor outcomes. Such unexpected adverse events are termed hyperprogressive disease (HPD), and their occurrence suggests that ICIs are detrimental to a subset of cancer patients. HPD is common, with an incidence ranging between 4 and 29% in several cancer types. However, the mechanisms of HPD remain poorly understood, and no clinical predictive factors of HPD have been identified. In this review, we summarize current findings, including retrospective studies and case reports, and focus on several key issues including the defining characteristics, predictive biomarkers, potential mechanisms of HPD, and strategies for avoiding HPD after ICI treatment.
Collapse
Affiliation(s)
- Pan Shen
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Liang Han
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Xin Ba
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Kai Qin
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| | - Shenghao Tu
- Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College of Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
111
|
Lin M, Vanneste BGL, Yu Q, Chen Z, Peng J, Cai X. Hyperprogression under immunotherapy: a new form of immunotherapy response?-a narrative literature review. Transl Lung Cancer Res 2021; 10:3276-3291. [PMID: 34430364 PMCID: PMC8350090 DOI: 10.21037/tlcr-21-575] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/26/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Update the last known review, and summarize the definitions, diagnostic criteria, reported risk factors, possible mechanisms and potential biomarkers of hyperprogressive disease (HPD) under immunotherapy. BACKGROUND Immunotherapy is a relatively new systemic therapy adding a new method of treatment of especially advanced cancer patients. In a variety of immunotherapies, however, an unexpected acceleration of tumor growth, known as HPD, is observed in approximately 30% of patients after immune checkpoint inhibitor (ICI) treatment. HPD has a deleterious survival effect on patients and represents an urgent issue for both clinicians and patients. Existing literature has reviewed and summarized the definition, diagnostic criteria, reported risk factors and possible mechanisms of hyperprogression. However, with the gradual deepening of the exploration of HPD, researchers have made significant breakthroughs in elucidating the mechanism and mechanism of HPD and exploring biomarkers. METHODS The search was conducted on Google Scholar and PubMed in January and May of 2021. We searched among English papers with no limitation on the publication year. We have included retrospective studies, case reports and basic researches related to HPD in the collection, we also referred to some review articles on HPD in recent years. A qualitative-interpretive approach was used for data extraction. CONCLUSIONS HPD is considered to be an acceleration of tumor growth after ICI treatment that is not only due to immune infiltration but also due to real disease progression, with an incidence of about 4-30% in all retrospective published studies to date. Currently, the most widely used criteria of HPD contain Response Evaluation Criteria in Solid Tumors (RECIST) and tumor growth rate (TGR) or tumor growth kinetics. The common risk factors and underlying mechanisms of HPD have not yet been fully elucidated. However, based on the poor prognosis of HPD, there have been many advances in the exploration of biomarkers in recent years, like the prediction of HPD, such as LDH levels of peripheral blood, liquid biopsy, and radiomics, etc.
Collapse
Affiliation(s)
- Miaozhen Lin
- Department of VIP Impatient, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ben G. L. Vanneste
- Department of Radiation Oncology (MAASTRO Clinic), GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Qiwen Yu
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Zebin Chen
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Jiayu Peng
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiuyu Cai
- Department of VIP Impatient, Sun Yat-sen University Cancer Center, Guangzhou, China
| |
Collapse
|
112
|
Selecting the optimal immunotherapy regimen in driver-negative metastatic NSCLC. Nat Rev Clin Oncol 2021; 18:625-644. [PMID: 34168333 DOI: 10.1038/s41571-021-00520-1] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2021] [Indexed: 12/12/2022]
Abstract
The treatment landscape of driver-negative non-small-cell lung cancer (NSCLC) is rapidly evolving. Immune-checkpoint inhibitors, specifically those targeting PD-1 or PD-L1, have demonstrated durable efficacy in a subset of patients with NSCLC, and these agents have become the cornerstone of first-line therapy. Approved immunotherapeutic strategies for treatment-naive patients now include monotherapy, immunotherapy-exclusive regimens or chemotherapy-immunotherapy combinations. Decision making in this space is complex given the absence of head-to-head prospective comparisons, although a thorough analysis of long-term efficacy and safety data from pivotal clinical trials can provide insight into the optimal management of each subset of patients. Indeed, histological subtype and the extent of tumour cell PD-L1 expression are paramount to regimen selection, although other clinicopathological factors and patient preferences might also be relevant in certain scenarios. Finally, several emerging biomarkers and novel therapeutic strategies are currently under investigation, and these might further refine the current treatment paradigm. In this Review, we discuss the current treatment landscape and detail our approach to first-line immunotherapy regimen selection for patients with advanced-stage, driver-negative NSCLC.
Collapse
|
113
|
Hyperprogressive Disease Caused by PD-1 Inhibitors for the Treatment of Pan-Cancer. DISEASE MARKERS 2021; 2021:6639366. [PMID: 34239621 PMCID: PMC8241516 DOI: 10.1155/2021/6639366] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 06/02/2021] [Indexed: 12/25/2022]
Abstract
Background Nowadays, PD-1/PD-L1 inhibitors are widely used to treat various malignant tumors. However, during the immunotherapy in a few patients, a flare-up of tumor growth occurred. This new pattern of progression is called hyperprogressive disease (HPD). Patients and Methods. The retrospective study included 377 patients with various malignant tumors treated with PD-1 inhibitors (nivolumab or pembrolizumab) in the Chinese PLA General Hospital from January 2015 to January 2019. Clinicopathologic variables, tumor growth rate (TGR), and treatment outcomes were analyzed in patients with pan-cancer treated with PD-1 inhibitors. HPD was defined as the difference of TGR before and during immunotherapy exceeding 50%. Results In 38 of 377 patients (10.08%), HPD occurred after treatment with PD-1 inhibitors. Patients with HPD had lower overall survival (OS) than those without HPD (median OS, 3.6months (95% CI, 3.0-4.2) vs. 7.3 months (95% CI, 5.9-8.7); P < 0.01). Factors related to HPD include more than 2 metastatic sites, ECOG performance status ≥ 2, hepatic metastases, and lactate dehydrogenase level greater than normal upper limit. KRAS status was significantly associated with HPD in patients with colorectal cancer. In the exploratory predictors' analysis, the rapid increase of characteristic tumor markers (such as CEA in colorectal cancer, CA199 in pancreatic cancer and cholangiocarcinoma) within one month was found to be associated with the occurrence of HPD. Conclusions HPD was developed with different rates in a variety of malignant tumor patients treated with PD-1 inhibitors and related to some clinicopathological features and poor prognosis. Tumor markers, especially CA199, might be served as early predictors of HPD.
Collapse
|
114
|
Wu M, Liu J, Seery S, Meng X, Yue J. Cytoreductive Nephrectomy Promoted Abscopal Effect of Camrelizumab Combined With Radiotherapy for Metastatic Renal Cell Carcinoma: A Case Report and Review of the Literature. Front Immunol 2021; 12:646085. [PMID: 34211459 PMCID: PMC8239433 DOI: 10.3389/fimmu.2021.646085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 05/28/2021] [Indexed: 11/13/2022] Open
Abstract
There is little evidence around Camrelizumab combined with cytoreductive nephrectomy (CN) and radiotherapy (RT) as a treatment option for metastatic renal cell carcinoma (mRCC). The influence of CN on immune responses and the abscopal effect are not well understood. In this paper, we report a case of anti-programmed cell death-1 (PD-1) treated with combined RT once CN reduced the primary tumor burden (TB). This patient also encountered an increased response to targeted radiotherapy after immune resistance. We also observed a macrophage-to-lymphocyte ratio (MLR) peak, which may be correlated with subsequent pseudoprogression after thoracic radiotherapy. Consequently, even with the disease, this patient has remained stable. This peculiar instance suggests there is a need to investigate the underlying mechanisms of CN in promoting the abscopal effect during immunotherapy when combined with RT. It also suggests that there is a need for further investigation into the role of RT in overcoming immune resistance, and the value of MLR in predicting pseudoprogression. We hypothesize that a heavy tumor burden might suppress the abscopal effect, thereby ensuring that CN promotes it. However, radiotherapy may overcome immune resistance during oligoprogression.
Collapse
Affiliation(s)
- Min Wu
- Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jie Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Samuel Seery
- Health Research, Faculty of Health and Medicine, Lancaster University, City of Lancaster, United Kingdom
| | - Xue Meng
- Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jinbo Yue
- Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
115
|
Ferrara R, Signorelli D, Proto C, Prelaj A, Garassino MC, Lo Russo G. Novel patterns of progression upon immunotherapy in other thoracic malignancies and uncommon populations. Transl Lung Cancer Res 2021; 10:2955-2969. [PMID: 34295690 PMCID: PMC8264338 DOI: 10.21037/tlcr-20-636] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 10/21/2020] [Indexed: 11/06/2022]
Abstract
In the immunotherapy era, considering the prolonged survival benefit and responses observed with immunecheckpoint inhibitors (ICI) in many cancer types, the identification of patients with rapid progression (PD) and deaths upon ICI has found some skepticism and resistance among the scientific community. Nevertheless, an acceleration of tumour during ICI, defined as hyperprogressive disease (HPD), has been recognized across different cancer types and evidence regarding rapid PDs and deaths are emerging in patients with malignant pleural mesothelioma (MPM), small cell lung cancer (SCLC) and thymic malignancies and in uncommon non-small cell lung cancer (NSCLC) populations. Of note, PD and early deaths (ED) rates upon single agent ICI were up to 60% and 30% in MPM and 70% and 38% in SCLC patients, respectively. Similarly, rapid PDs and deaths were observed in clinical trials and retrospective studies including patients with poor performance status (PS), HIV infection and rare NSCLC histologies. Atypical patterns of response, such as pseudoprogression (PsPD) may also occur in other thoracic malignancies (MPM) and in some uncommon populations (i.e., HIV patients), however probably at lower rate compared to HPD. The characterizations of HPD and PsPD mechanisms and the identification of common definition criteria are the next future challenges in this area of cancer research.
Collapse
Affiliation(s)
- Roberto Ferrara
- Department of Medical Oncology, Thoracic Oncology Unit, Fondazione IRCSS, Istituto Nazionale dei Tumori Milano, Milan, Italy.,Department of Research, Molecular Immunology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Diego Signorelli
- Department of Medical Oncology, Thoracic Oncology Unit, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Claudia Proto
- Department of Medical Oncology, Thoracic Oncology Unit, Fondazione IRCSS, Istituto Nazionale dei Tumori Milano, Milan, Italy
| | - Arsela Prelaj
- Department of Medical Oncology, Thoracic Oncology Unit, Fondazione IRCSS, Istituto Nazionale dei Tumori Milano, Milan, Italy.,Department of Electronics, Information, and Bioengineering, Polytechnic University of Milan, Milan, Italy
| | - Marina Chiara Garassino
- Department of Medical Oncology, Thoracic Oncology Unit, Fondazione IRCSS, Istituto Nazionale dei Tumori Milano, Milan, Italy
| | - Giuseppe Lo Russo
- Department of Medical Oncology, Thoracic Oncology Unit, Fondazione IRCSS, Istituto Nazionale dei Tumori Milano, Milan, Italy
| |
Collapse
|
116
|
Feng D, Guan Y, Liu M, He S, Zhao W, Yin B, Liang J, Li Y, Wang J. Excellent Response to Atezolizumab After Clinically Defined Hyperprogression Upon Previous Treatment With Pembrolizumab in Metastatic Triple-Negative Breast Cancer: A Case Report and Review of the Literature. Front Immunol 2021; 12:608292. [PMID: 34135884 PMCID: PMC8201609 DOI: 10.3389/fimmu.2021.608292] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 05/07/2021] [Indexed: 12/17/2022] Open
Abstract
Immunotherapy with immune checkpoint inhibitors (ICIs), including programmed cell death protein-1 (PD-1) and programmed cell death ligand-1 (PD-L1) inhibitors, has revolutionized the systematic treatment of advanced and metastatic solid tumors. However, the response rate to ICIs is unsatisfactory, and unexpected hyperprogressive disease (HPD) is even observed in a small subgroup of patients. Patients with HPD usually have worsening clinical symptoms and poorer survival, and therapeutic strategies are extremely limited. Here, we presented a patient with HPD who had used a PD-L1 inhibitor and was highly responsive to the sequential use of a PD-1 inhibitor. A 67-year-old woman with metastatic triple-negative breast cancer was treated with pembrolizumab plus chemotherapy after progression on previous multiple-line chemotherapy treatments. After 2 cycles of treatments, she rapidly developed HPD, as confirmed by radiological evaluation and worsening symptoms. At that time, pembrolizumab was discontinued, and she switched to the PD-L1 inhibitor atezolizumab plus chemotherapy. This patient partially responded to atezolizumab plus chemotherapy without experiencing severe drug-related adverse effects. This is the first reported case of metastatic breast cancer in a patient with radiologically confirmed HPD after pembrolizumab therapy in which successful rechallenge with atezolizumab relieved clinical symptoms. Further studies with larger sample sizes involving a deeper translational investigation of HPD are needed to confirm the efficacy and mechanism of sequential application of different ICIs for the clinical management of HPD.
Collapse
Affiliation(s)
- Dongfeng Feng
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.,Shandong Lung Cancer Institute, Jinan, China.,Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Yaping Guan
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.,Shandong Lung Cancer Institute, Jinan, China.,Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Mingguo Liu
- Department of Oncology, Yuncheng Honesty Hospital, Heze, China
| | - Shuqian He
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University, Jinan, China
| | - Weipeng Zhao
- Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Beibei Yin
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.,Shandong Lung Cancer Institute, Jinan, China.,Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Jing Liang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.,Shandong Lung Cancer Institute, Jinan, China.,Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Yan Li
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.,Shandong Lung Cancer Institute, Jinan, China.,Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| | - Jun Wang
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China.,Shandong Lung Cancer Institute, Jinan, China.,Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, China
| |
Collapse
|
117
|
Gkountakos A, Delfino P, Lawlor RT, Scarpa A, Corbo V, Bria E. Harnessing the epigenome to boost immunotherapy response in non-small cell lung cancer patients. Ther Adv Med Oncol 2021; 13:17588359211006947. [PMID: 34104224 PMCID: PMC8161860 DOI: 10.1177/17588359211006947] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 03/11/2021] [Indexed: 12/13/2022] Open
Abstract
The introduction of immune checkpoint inhibitor (ICI)-based therapy for non-oncogene addicted non-small cell lung cancer (NSCLC) has significantly transformed the treatment landscape of the disease. Inhibitors of the programmed cell death protein 1/programmed death-ligand 1 (PD-1/PD-L1) immune checkpoint axis, which were initially considered as a late-line treatment option, gradually became the standard of care as first-line treatment for subgroups of NSCLC patients. However, a significant fraction of patients either fails to respond or progresses after a partial response to ICI treatment. Thus, the identification of mechanisms responsible for innate and acquired resistance to immunotherapy within a rapidly evolving tumor microenvironment (TME) is urgently required, as is the identification of reliable predictive biomarkers beyond PD-L1 expression. The deregulation of the epigenome is a key driver of cancer initiation and progression, and it has also been shown to drive therapeutic resistance. Tumor education of infiltrating myeloid cells towards an immuno-suppressive phenotype as well as induction of T-cell dysfunction in the TME is also driven by epigenome reprogramming. As it stands and, given their dynamic nature, epigenetic changes in cancer and non-cancer cells represent an attractive target to increase immunotherapy activity in NSCLC. Accordingly, clinical trials of combinatorial immuno-epigenetic drug regimens have been associated with tumor response in previously immunotherapy-resistant NSCLC patients irrespective of their PD-L1 status. Moreover, epigenetic signatures might represent valuable theragnostic biomarkers as they can be assayed easily in liquid biopsy and provide multiple layers of information. In this review, we discuss the current knowledge regarding the dysregulated epigenetic mechanisms contributing to immunotherapy resistance in NSCLC. Although the clinical data are still maturing, we highlight the attractive perspective that the synergistic model of immuno-epigenetic strategies might overcome the current limitations of immunotherapy alone and will be translated into durable clinical benefit for a broader NSCLC population.
Collapse
Affiliation(s)
- Anastasios Gkountakos
- ARC-NET Applied Research on Cancer Center, University of Verona, P.le L.A. Scuro 10, Verona, 37134, Italy
| | - Pietro Delfino
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Rita T. Lawlor
- ARC-NET Applied Research on Cancer Center, University of Verona, Verona, Italy
| | - Aldo Scarpa
- ARC-NET Applied Research on Cancer Center, University of Verona, Verona, Italy
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Vincenzo Corbo
- ARC-NET Applied Research on Cancer Center, University of Verona, Verona, Italy
- Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | - Emilio Bria
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Department of Medical Oncology, Università Cattolica Del Sacro Cuore, Rome, Italy
| |
Collapse
|
118
|
Ippolito D, Maino C, Ragusi M, Porta M, Gandola D, Franzesi CT, Giandola TP, Sironi S. Immune response evaluation criteria in solid tumors for assessment of atypical responses after immunotherapy. World J Clin Oncol 2021; 12:323-334. [PMID: 34131564 PMCID: PMC8173324 DOI: 10.5306/wjco.v12.i5.323] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/23/2021] [Accepted: 04/23/2021] [Indexed: 02/06/2023] Open
Abstract
In 2017, immune response evaluation criteria in solid tumors (iRECIST) were introduced to validate radiologic and clinical interpretations and to better analyze tumor’s response to immunotherapy, considering the different time of following and response, between this new therapy compared to the standard one. However, even if the iRECIST are worldwide accepted, to date, different aspects should be better underlined and well reported, especially in clinical practice. Clinical experience has demonstrated that in a non-negligible percentage of patients, it is challenging to determine the correct category of response (stable disease, progression disease, partial or complete response), and consequently, to define which is the best management for those patients. Approaching radiological response in patients who underwent immunotherapy, a new uncommon kind of target lesions behavior was found. This phenomenon is mainly due to the different mechanisms of action of immunotherapeutic drug. Therefore, new groups of response have been described in clinical practice, defined as “atypical responses,” and categorized into three new groups: pseudoprogression, hyperprogression, and dissociated response. This review summarizes and reports these patterns, helping clinicians and radiologists get used to atypical responses, in order to identify patients that respond best to treatment.
Collapse
Affiliation(s)
- Davide Ippolito
- Department of Diagnostic Radiology, H. S. Gerardo Monza, School of Medicine, University of Milano-Bicocca, Monza 20900, Italy
| | - Cesare Maino
- Department of Diagnostic Radiology, H. S. Gerardo Monza, School of Medicine, University of Milano-Bicocca, Monza 20900, Italy
| | - Maria Ragusi
- Department of Diagnostic Radiology, H. S. Gerardo Monza, School of Medicine, University of Milano-Bicocca, Monza 20900, Italy
| | - Marco Porta
- Department of Diagnostic Radiology, H. S. Gerardo Monza, School of Medicine, University of Milano-Bicocca, Monza 20900, Italy
| | - Davide Gandola
- Department of Diagnostic Radiology, H. S. Gerardo Monza, School of Medicine, University of Milano-Bicocca, Monza 20900, Italy
| | - Cammillo Talei Franzesi
- Department of Diagnostic Radiology, H. S. Gerardo Monza, School of Medicine, University of Milano-Bicocca, Monza 20900, Italy
| | - Teresa Paola Giandola
- Department of Diagnostic Radiology, H. S. Gerardo Monza, School of Medicine, University of Milano-Bicocca, Monza 20900, Italy
| | - Sandro Sironi
- Diagnostic Radiology, University of Milano-Bicocca, Bergamo 24127, Italy
| |
Collapse
|
119
|
Zhou K, Sun M, Xia Y, Xie Y, Shu R. LPS stimulates gingival fibroblasts to express PD-L1 via the p38 pathway under periodontal inflammatory conditions. Arch Oral Biol 2021; 129:105161. [PMID: 34090065 DOI: 10.1016/j.archoralbio.2021.105161] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 02/04/2023]
Abstract
OBJECTIVE The overall aim of this research was to investigate the differences in the expression of programmed death ligand 1 (PD-L1) in human gingival fibroblasts (HGFs) between a periodontal healthy group and a periodontal inflammatory group. and explore the possible mechanism involved. METHODS Differences in PD-L1 mRNA and protein expression in HGFs from a periodontal healthy group and a periodontal inflammatory group were examined by qPCR and western blotting, respectively, and were further tested after lipopolysaccharide (LPS) stimulation in both groups. The effects of a p38 pathway inhibitor on the changes in p38 phosphorylation levels and PD-L1 expression after LPS stimulation were investigated in both groups. RESULTS PD-L1 mRNA and protein levels in HGFs in the periodontal inflammatory group were significantly higher than those in the periodontal healthy group (p < 0.05). After 10 μg/mL LPS stimulation, PD-L1 mRNA levels in HGFs from both groups increased significantly (p < 0.05), peaking at 4 h, and the peak was significantly higher in the periodontal inflammatory group than in the periodontal healthy group (p < 0.05). However, PD-L1 protein expression was upregulated only in the inflammatory group (p < 0.05). Inhibition of the p38 pathway in HGFs decreased p38 phosphorylation in both groups (p < 0.05) but this treatment reversed the LPS-induced increase in PD-L1 mRNA and protein levels only in the inflammatory group (p < 0.05). CONCLUSION In the periodontal inflammatory state, the expression of PD-L1 in HGFs is more easily activated, and may be influenced by the p38 pathway.
Collapse
Affiliation(s)
- Kecong Zhou
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, Shanghai, China; National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology, Shanghai, China; Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital Research Center, Shanghai Jiao Tong University, Shanghai, China
| | - Mengjun Sun
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, Shanghai, China; National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology, Shanghai, China; Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital Research Center, Shanghai Jiao Tong University, Shanghai, China
| | - Yiru Xia
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, Shanghai, China; National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology, Shanghai, China; Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital Research Center, Shanghai Jiao Tong University, Shanghai, China
| | - Yufeng Xie
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, Shanghai, China; National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology, Shanghai, China; Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital Research Center, Shanghai Jiao Tong University, Shanghai, China.
| | - Rong Shu
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; College of Stomatology, Shanghai Jiao Tong University, Shanghai, China; National Center for Stomatology, Shanghai, China; National Clinical Research Center for Oral Diseases, Shanghai, China; Shanghai Key Laboratory of Stomatology, Shanghai, China; Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital Research Center, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
120
|
Biomarkers or factors for predicting the efficacy and adverse effects of immune checkpoint inhibitors in lung cancer: achievements and prospective. Chin Med J (Engl) 2021; 133:2466-2475. [PMID: 32960841 PMCID: PMC7575173 DOI: 10.1097/cm9.0000000000001090] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are widely used in lung cancer therapy due to their effectiveness and minimal side effects. However, only a few lung cancer patients benefit from ICI therapy, driving the need to develop alternative biomarkers. Programmed death-ligand 1 (PD-L1) molecules expressed in tumor cells and immune cells play a key role in the immune checkpoint pathway. Therefore, PD-L1 expression is a prognostic biomarker in evaluating the effectiveness of programmed death-1 (PD-1)/PD-L1 inhibitors. Nevertheless, adverse predictive outcomes suggest that other factors are implicated in the response. In this review, we present a detailed introduction of existing biomarkers concerning tumor abnormality and host immunity. PD-L1 expression, tumor mutation burden, neoantigens, specific gene mutations, circulating tumor DNA, human leukocyte antigen class I, tumor microenvironment, peripheral inflammatory cells, and microbiome are discussed in detail. To sum up, this review provides information on the current application and future prospects of ICI biomarkers.
Collapse
|
121
|
Ayers KL, Mullaney T, Zhou X, Liu JJ, Lee K, Ma M, Jones S, Li L, Redfern A, Jappe W, Liu Z, Goldsweig H, Yadav KK, Hahner N, Dietz M, Zimmerman M, Prentice T, Newman S, Veluswamy R, Wisnivesky J, Hirsch FR, Oh WK, Li SD, Schadt EE, Chen R. Analysis of Real-World Data to Investigate the Impact of Race and Ethnicity on Response to Programmed Cell Death-1 and Programmed Cell Death-Ligand 1 Inhibitors in Advanced Non-Small Cell Lung Cancers. Oncologist 2021; 26:e1226-e1239. [PMID: 33829580 PMCID: PMC8265370 DOI: 10.1002/onco.13780] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 02/25/2021] [Indexed: 12/19/2022] Open
Abstract
Background Racial disparities among clinical trial participants present a challenge to assess whether trial results can be generalized into patients representing diverse races and ethnicities. The objective of this study was to evaluate the impact of race and ethnicity on treatment response in patients with advanced non‐small cell lung cancer (aNSCLC) treated with programmed cell death‐1 (PD‐1) or programmed cell death‐ligand 1 (PD‐L1) inhibitors through analysis of real‐world data (RWD). Materials and Methods A retrospective cohort study of 11,138 patients with lung cancer treated at hospitals within the Mount Sinai Health System was performed. Patients with confirmed aNSCLC who received anti‐PD‐1/PD‐L1 treatment were analyzed for clinical outcomes. Our cohort included 249 patients with aNSCLC who began nivolumab, pembrolizumab, or atezolizumab treatment between November 2014 and December 2018. Time‐to‐treatment discontinuation (TTD) and overall survival (OS) were the analyzed clinical endpoints. Results After a median follow‐up of 14.8 months, median TTD was 7.8 months (95% confidence interval, 5.4–not estimable [NE]) in 75 African American patients versus 4.6 (2.4–7.2) in 110 White patients (hazard ratio [HR], 0.63). Median OS was not reached (18.4–NE) in African American patients versus 11.6 months (9.7–NE) in White patients (HR, 0.58). Multivariable Cox regression conducted with potential confounders confirmed longer TTD (adjusted HR, 0.65) and OS (adjusted HR, 0.60) in African American versus White patients. Similar real‐world response rate (42.6% vs. 43.5%) and disease control rate (59.6% vs. 56.5%) were observed in the African American and White patient populations. Further investigation revealed the African American patient group had lower incidence (14.7%) of putative hyperprogressive diseases (HPD) upon anti‐PD‐1/PD‐L1 treatment than the White patient group (24.5%). Conclusion Analysis of RWD showed longer TTD and OS in African American patients with aNSCLC treated with anti‐PD‐1/PD‐L1 inhibitors. Lower incidence of putative HPD is a possible reason for the favorable outcomes in this patient population. Implications for Practice There is a significant underrepresentation of minority patients in randomized clinical trials, and this study demonstrates that real‐world data can be used to investigate the impact of race and ethnicity on treatment response. In retrospective analysis of patients with advanced non‐small cell lung cancer treated with programmed cell death‐1 or programmed cell death‐ligand 1 inhibitors, African American patients had significantly longer time‐to‐treatment discontinuation and longer overall survival. Analysis of real‐world data can yield clinical insights and establish a more complete picture of medical interventions in routine clinical practice. Racial disparities in clinical trials affect whether trial results can be generalized for diverse patient populations. This article evaluates real‐world data to assess the impact of race and ethnicity on treatment response in patients with advanced non‐small cell lung cancer treated with PD‐1 or PD‐L1 inhibitors.
Collapse
Affiliation(s)
| | | | | | - Jane J Liu
- Sema4, Stamford, Connecticut, USA.,Illinois CancerCare, Peoria, Illinois, USA
| | | | - Meng Ma
- Sema4, Stamford, Connecticut, USA
| | | | - Li Li
- Sema4, Stamford, Connecticut, USA.,Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Juan Wisnivesky
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Fred R Hirsch
- Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - William K Oh
- Sema4, Stamford, Connecticut, USA.,Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shuyu D Li
- Sema4, Stamford, Connecticut, USA.,Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Eric E Schadt
- Sema4, Stamford, Connecticut, USA.,Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rong Chen
- Sema4, Stamford, Connecticut, USA.,Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
122
|
Li J, Xiang C, Wang Y, Zhou Y, Cao S, Ling X, Ye J, Zheng J, Shao L, Zhong H, Han Y. The genomic characteristics of different progression patterns in advanced non-small cell lung cancer patients treated with immune checkpoint inhibitors. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:779. [PMID: 34268392 PMCID: PMC8246171 DOI: 10.21037/atm-20-6910] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/04/2021] [Indexed: 02/04/2023]
Abstract
Background Fast progression (FP), hyperprogressive disease (HPD), and early death (ED) are the newly reported cancer progression patterns in response to immune checkpoint inhibitor (ICI) treatment. This study aimed to investigate the clinical and genomic characteristics of FP, HPD, and ED following the ICI treatment of advanced non-small cell lung cancer (NSCLC). Methods We retrospectively reviewed 117 patients with advanced NSCLC who were treated with ICIs from March 2017 to October 2019. FP was defined as (I) time to treatment failure (TTF) <1.5 months; and (II) ≥50% increase in the sum of the longest diameter (SLD) of target lesions. HPD was defined as (I) TTF <2 months; and (II) ≥50% change in tumor growth rate compared with before ICI initiation. ED was defined as overall survival (OS) <3 months. Tissue samples from 18 FP/HPD/ED patients and 5 partial response (PR) patients were subjected to genomic profiling. Genomic data from 693 tumor mutational burden- and histology-matched lung cancer samples were retrieved from an internal database as a control. Results FP, HPD, and ED occurred in 7.21%, 9.38%, and 11.97% patients, respectively. The progression-free survival was comparable among the 3 groups. The median overall survival for FP, HPD, and ED were 3.19, 11.2, and 1.84 months, respectively. The genomic landscape revealed 1 EGFR amplification, 1 ALK fusion, 6 KRAS mutations, 1 ERBB2 amplification, 1 MET amplification, and 1 RET fusion among the 18 patients with FP/HPD/ED. Compared with the Control group, ED patients showed higher mutation frequencies for KRAS (P<0.01), CDKN1B (P<0.01), and NTRK1 (P=0.04). Mutations in RAD54L (P=0.018) and MYC (P=0.04) were more common in FP patients; HPD patients showed more frequent RAD54L mutations (P<0.001). Conclusions We demonstrated different genomic characteristics across different progression patterns following ICI treatment, which might assist clinicians in the prediction of a patient’s response, identifying candidates for more effective ICI therapy.
Collapse
Affiliation(s)
- Jingwen Li
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Chan Xiang
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yue Wang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yan Zhou
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shuhui Cao
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xuxinyi Ling
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Junyi Ye
- Department of Medicine and Clinical Research, Burning Rock Biotech, Guangzhou, China
| | - Jingjing Zheng
- Department of Medicine and Clinical Research, Burning Rock Biotech, Guangzhou, China
| | - Lin Shao
- Department of Medicine and Clinical Research, Burning Rock Biotech, Guangzhou, China
| | - Hua Zhong
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yuchen Han
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
123
|
Ayers KL, Ma M, Debussche G, Corrigan D, McCafferty J, Lee K, Newman S, Zhou X, Hirsch FR, Mack PC, Liu JJ, Schadt EE, Chen R, Li SD. A composite biomarker of neutrophil-lymphocyte ratio and hemoglobin level correlates with clinical response to PD-1 and PD-L1 inhibitors in advanced non-small cell lung cancers. BMC Cancer 2021; 21:441. [PMID: 33882890 PMCID: PMC8059160 DOI: 10.1186/s12885-021-08194-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/13/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have been incorporated into various clinical oncology guidelines for systemic treatment of advanced non-small cell lung cancers (aNSCLC). However, less than 50% (and 20%) of the patients responded to the therapy as a first (or second) line of therapy. PD-L1 immunohistochemistry (IHC) is an extensively studied biomarker of response to ICI, but results from this test have equivocal predictive power. In order to identify other biomarkers that support clinical decision-making around whether to treat with ICIs or not, we performed a retrospective study of patients with aNSCLC who underwent ICI-based therapy in the Mount Sinai Health System between 2014 and 2019. METHODS We analyzed data from standard laboratory tests performed in patients as a part of the routine clinical workup during treatment, including complete blood counts (CBC) and a comprehensive metabolic panel (CMP), to correlate test results with clinical response and survival. RESULTS Of 11,138 NSCLC patients identified, 249 had been treated with ICIs. We found associations between high neutrophil-to-lymphocyte ratio (NLR ≥ 5) and poor survival in ICI-treated NSCLC. We further observed that sustained high NLR after initiation of treatment had a more profound impact on survival than baseline NLR, regardless of PD-L1 status. Hazard ratios when comparing patients with NLR ≥ 5 vs. NLR < 5 are 1.7 (p = 0.02), 3.4 (p = 4.2 × 10- 8), and 3.9 (p = 1.4 × 10- 6) at baseline, 2-8 weeks, and 8-14 weeks after treatment start, respectively. Mild anemia, defined as hemoglobin (HGB) less than 12 g/dL was correlated with survival independently of NLR. Finally, we developed a composite NLR and HGB biomarker. Patients with pretreatment NLR ≥ 5 and HGB < 12 g/dL had a median overall survival (OS) of 8.0 months (95% CI 4.5-11.5) compared to the rest of the cohort with a median OS not reached (95% CI 15.9-NE, p = 1.8 × 10- 5), and a hazard ratio of 2.6 (95% CI 1.7-4.1, p = 3.5 × 10- 5). CONCLUSIONS We developed a novel composite biomarker for ICI-based therapy in NSCLC based on routine CBC tests, which may provide meaningful clinical utility to guide treatment decision. The results suggest that treatment of anemia to elevate HGB before initiation of ICI therapy may improve patient outcomes or the use of alternative non-chemotherapy containing regimens.
Collapse
Affiliation(s)
- Kristin L Ayers
- Sema4, a Mount Sinai Venture, 333 Ludlow Street, Stamford, CT, 06902, USA
| | - Meng Ma
- Sema4, a Mount Sinai Venture, 333 Ludlow Street, Stamford, CT, 06902, USA
| | - Gaspard Debussche
- Sema4, a Mount Sinai Venture, 333 Ludlow Street, Stamford, CT, 06902, USA
| | - David Corrigan
- Sema4, a Mount Sinai Venture, 333 Ludlow Street, Stamford, CT, 06902, USA
| | | | - Kyeryoung Lee
- Sema4, a Mount Sinai Venture, 333 Ludlow Street, Stamford, CT, 06902, USA
| | - Scott Newman
- Sema4, a Mount Sinai Venture, 333 Ludlow Street, Stamford, CT, 06902, USA
| | - Xiang Zhou
- Sema4, a Mount Sinai Venture, 333 Ludlow Street, Stamford, CT, 06902, USA
| | - Fred R Hirsch
- Center of Thoracic Oncology/Tisch Cancer Institute and Icahn School of Medicine, Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Philip C Mack
- Center of Thoracic Oncology/Tisch Cancer Institute and Icahn School of Medicine, Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Jane J Liu
- Sema4, a Mount Sinai Venture, 333 Ludlow Street, Stamford, CT, 06902, USA.,Illinois Cancer Care, 8940 N Wood Sage Rd, Peoria, IL, 61615, USA
| | - Eric E Schadt
- Sema4, a Mount Sinai Venture, 333 Ludlow Street, Stamford, CT, 06902, USA. .,Center of Thoracic Oncology/Tisch Cancer Institute and Icahn School of Medicine, Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA.
| | - Rong Chen
- Sema4, a Mount Sinai Venture, 333 Ludlow Street, Stamford, CT, 06902, USA. .,Center of Thoracic Oncology/Tisch Cancer Institute and Icahn School of Medicine, Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA.
| | - Shuyu D Li
- Sema4, a Mount Sinai Venture, 333 Ludlow Street, Stamford, CT, 06902, USA. .,Center of Thoracic Oncology/Tisch Cancer Institute and Icahn School of Medicine, Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA.
| |
Collapse
|
124
|
Liu J, Wu Q, Wu S, Xie X. Investigation on potential biomarkers of hyperprogressive disease (HPD) triggered by immune checkpoint inhibitors (ICIs). Clin Transl Oncol 2021; 23:1782-1793. [PMID: 33847923 DOI: 10.1007/s12094-021-02579-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/01/2021] [Indexed: 02/04/2023]
Abstract
PURPOSE This project aimed to survey the clinical characteristics and survivals of hyperprogressive disease (HPD) mediated by immune checkpoint inhibitors (ICIs) in an attempt to explore the potential predictors. METHODS After searching PubMed, MEDLINE, Google Scholar and Cochrane Library databases, 12 studies incorporating 1766 individuals were enrolled. The data were analyzed with Review manager 5.3 software. RESULTS The results revealed HPD correlated with previous metastatic sites > 2 (OR = 1.86, 95% CI 1.33-2.59, P = 0.0003), liver metastasis (OR = 3.35, 95% CI 2.09-5.35, P < 0.00001), Royal Marsden Hospital (RMH) score ≥ 2 (OR = 2.80, 95% CI 1.85-4.23, P < 0.00001), higher ECOG PS (OR = 1.60, 95% CI 1.13-2.27, P = 0.008) and LDH > upper limits of normal (ULN) (OR = 2.32, 95% CI 1.51-3.58, P = 0.0001). Instead, HPD was unrelated to gender, age, smoking status, PD-L1 expression, therapy, neutrophil-to-lymphocyte ratio, the histology, the status of EGFR, ALK and KRAS in non-small cell lung cancer and HER-2 expression in advanced gastric cancer. Moreover, HPD was evidently correlated with a shorter OS (HR = 2.92, 95% CI 1.79-4.76, P < 0.0001) and PFS (HR = 3.62, 95% CI 2.79-4.68, P < 0.00001). The same phenomena existed in stratified studies based on study regions and tumor types. CONCLUSIONS This study demonstrated that HPD was related to the number of prior metastatic sites > 2, liver metastasis, RMH score ≥ 2, higher ECOG PS score and LDH > ULN. Moreover, HPD was correlated with a poor OS and PFS in patients following ICI therapy.
Collapse
Affiliation(s)
- J Liu
- Department of Oncology, Molecular Oncology Research Institute, the First Affiliated Hospital, Fujian Medical University, Chazhong Road No 20, Fuzhou 350005, Fujian, China
| | - Q Wu
- Department of Oncology, Molecular Oncology Research Institute, the First Affiliated Hospital, Fujian Medical University, Chazhong Road No 20, Fuzhou 350005, Fujian, China
| | - S Wu
- Department of Oncology, Molecular Oncology Research Institute, the First Affiliated Hospital, Fujian Medical University, Chazhong Road No 20, Fuzhou 350005, Fujian, China
| | - X Xie
- Department of Oncology, Molecular Oncology Research Institute, the First Affiliated Hospital, Fujian Medical University, Chazhong Road No 20, Fuzhou 350005, Fujian, China. .,Fujian Key Laboratory of Precision Medicine for Cancer, the First Affiliated Hospital, Fujian Medical University, Chazhong Road No 20, Fuzhou 350005, Fujian, China.
| |
Collapse
|
125
|
Kwon M, An M, Klempner SJ, Lee H, Kim KM, Sa JK, Cho HJ, Hong JY, Lee T, Min YW, Kim TJ, Min BH, Park WY, Kang WK, Kim KT, Kim ST, Lee J. Determinants of Response and Intrinsic Resistance to PD-1 Blockade in Microsatellite Instability-High Gastric Cancer. Cancer Discov 2021; 11:2168-2185. [PMID: 33846173 DOI: 10.1158/2159-8290.cd-21-0219] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/28/2021] [Accepted: 04/08/2021] [Indexed: 11/16/2022]
Abstract
Sequence alterations in microsatellites and an elevated mutational burden are observed in 20% of gastric cancers and associated with clinical response to anti-PD-1 antibodies. However, 50% of microsatellite instability-high (MSI-H) cancers are intrinsically resistant to PD-1 therapies. We conducted a phase II trial of pembrolizumab in patients with advanced MSI-H gastric cancer and included serial and multi-region tissue samples in addition to serial peripheral blood analyses. The number of whole-exome sequencing (WES)-derived nonsynonymous mutations correlated with antitumor activity and prolonged progression-free survival (PFS). Coupling WES to single-cell RNA sequencing, we identified dynamic tumor evolution with greater on-treatment collapse of mutational architecture in responders. Diverse T-cell receptor repertoire was associated with longer PFS to pembrolizumab. In addition, an increase in PD-1+ CD8+ T cells correlated with durable clinical benefit. Our findings highlight the genomic, immunologic, and clinical outcome heterogeneity within MSI-H gastric cancer and may inform development of strategies to enhance responsiveness. SIGNIFICANCE: This study highlights response heterogeneity within MSI-H gastric cancer treated with pembrolizumab monotherapy and underscores the potential for extended baseline and early on-treatment biomarker analyses to identify responders. The observed markers of intrinsic resistance have implications for patient stratification to inform novel combinations among patients with intrinsically resistant features.See related commentary by Fontana and Smyth, p. 2126.This article is highlighted in the In This Issue feature, p. 2113.
Collapse
Affiliation(s)
- Minsuk Kwon
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Minae An
- Samsung Advanced Institute of Health Science and Technology, Sungkyunkwan University, Seoul, Korea
| | - Samuel J Klempner
- Department of Medicine, Division of Hematology-Oncology, Massachusetts General Hospital, Boston, Massachusetts
| | - Hyuk Lee
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyoung-Mee Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jason K Sa
- Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea
| | - Hee Jin Cho
- Innovative Institute for Precision Medicine, Samsung Medical Center, Seoul, Korea
| | - Jung Yong Hong
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Taehyang Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yang Won Min
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Tae Jun Kim
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Byung-Hoon Min
- Division of Gastroenterology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea
| | - Won Ki Kang
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyu-Tae Kim
- Department of Physiology, Ajou University School of Medicine, Suwon, Korea.
| | - Seung Tae Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Jeeyun Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea. .,Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Korea
| |
Collapse
|
126
|
Frelaut M, du Rusquec P, de Moura A, Le Tourneau C, Borcoman E. Pseudoprogression and Hyperprogression as New Forms of Response to Immunotherapy. BioDrugs 2021; 34:463-476. [PMID: 32394415 DOI: 10.1007/s40259-020-00425-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Indications of immunotherapy in oncology are continuously expanding, and unconventional types of response have been observed with these new treatments. These include transient progressive disease followed by a partial response, described as pseudoprogression, that raises the question of treatment beyond progression; and rapid disease progression associated with clinical decline, reported as hyperprogression. However, there are currently no consensual definitions of these phenomena and their impact on daily practice remains unclear. We reviewed existing data on pseudoprogression and hyperprogression with a focus on the definitions, incidence, predictive factors, potential biological mechanisms, and methods published to help distinguish pseudoprogression from progression and hyperprogression. The incidence of pseudoprogression ranged from 0 to 15%, with some authors also including disease stabilization after a first progression. For hyperprogression, incidence ranged from 4 to 29% with various definitions, and several authors reported a correlation with worse survival. Both phenomena were observed in a large panel of cancer types. Several radiological and biological methods have been reported to help distinguish pseudoprogression from progression and hyperprogression, such as analysis of radiomics, and circulating-tumor DNA or cell-free DNA, but these need to be confirmed in larger prospective cohorts. In conclusion, pseudoprogression and hyperprogression are both frequent types of responses under immunotherapy, and there is a need to better characterize these to improve the management of cancer patients. Treatment beyond progression should always be considered with caution and necessitates close clinical monitoring. In case of suspected hyperprogression, immunotherapy should be stopped early.
Collapse
Affiliation(s)
- Maxime Frelaut
- Department of Drug Development and Innovation (D3i), Institut Curie, Saint-Cloud, Paris, France
| | - Pauline du Rusquec
- Department of Drug Development and Innovation (D3i), Institut Curie, Saint-Cloud, Paris, France
| | - Alexandre de Moura
- Department of Drug Development and Innovation (D3i), Institut Curie, Saint-Cloud, Paris, France
| | - Christophe Le Tourneau
- Department of Drug Development and Innovation (D3i), Institut Curie, Saint-Cloud, Paris, France
- INSERM U900 Research Unit, Saint-Cloud, France
- Paris-Saclay University, Paris, France
| | - Edith Borcoman
- Department of Drug Development and Innovation (D3i), Institut Curie, Saint-Cloud, Paris, France.
| |
Collapse
|
127
|
Hyperprogressive Disease: Main Features and Key Controversies. Int J Mol Sci 2021; 22:ijms22073736. [PMID: 33916696 PMCID: PMC8038385 DOI: 10.3390/ijms22073736] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 03/31/2021] [Accepted: 03/31/2021] [Indexed: 12/29/2022] Open
Abstract
Along with the positioning of immunotherapy as a preferential treatment for a wide variety of neoplasms, a new pattern of response consisting in a sudden acceleration of tumor growth has been described. This phenomenon has received the name of "hyperprogressive disease", and several definitions have been proposed for its identification, most of them relying on radiological criteria. However, due to the fact that the cellular and molecular mechanisms have not been elucidated yet, there is still some debate regarding whether this fast progression is induced by immunotherapy or only reflects the natural course of some highly aggressive neoplasms. Moreover, contradictory results of trials including patients with different cancer types suggest that both the incidence, the associated factors and the implications regarding prognosis might differ depending on tumor histology. This article intends to review the main publications regarding this matter and critically approach the most controversial aspects.
Collapse
|
128
|
Sangaletti S, Ferrara R, Tripodo C, Garassino MC, Colombo MP. Myeloid cell heterogeneity in lung cancer: implication for immunotherapy. Cancer Immunol Immunother 2021; 70:2429-2438. [PMID: 33797567 PMCID: PMC8017108 DOI: 10.1007/s00262-021-02916-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 03/13/2021] [Indexed: 12/14/2022]
Abstract
Lung is a specialized tissue where metastases from primary lung tumors takeoff and those originating from extra-pulmonary sites land. One commonality characterizing these processes is the supportive role exerted by myeloid cells, particularly neutrophils, whose recruitment is facilitated in this tissue microenvironment. Indeed, neutrophils have important part in the pathophysiology of this organ and the key mechanisms regulating neutrophil expansion and recruitment during infection can be co-opted by tumor cells to promote growth and metastasis. Although neutrophils dominate the myeloid landscape of lung cancer other populations including macrophages, dendritic cells, mast cells, basophils and eosinophils contribute to the complexity of lung cancer TME. In this review, we discuss the origin and significance of myeloid cells heterogeneity in lung cancer, which translates not only in a different frequency of immune populations but it encompasses state of activation, morphology, localization and mutual interactions. The relevance of such heterogeneity is considered in the context of tumor growth and response to immunotherapy.
Collapse
Affiliation(s)
- Sabina Sangaletti
- Department of Research, Molecular Immunology Unit, Fondazione IRCCS Istituto Nazionale Dei Tumori, via Amadeo 42, 20133, Milano, Italy
| | - Roberto Ferrara
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Claudio Tripodo
- Tumor Immunology Unit, University of Palermo, Palermo, Italy.,FIRC Institute of Molecular Oncology (IFOM), Milano, Italy
| | - Marina Chiara Garassino
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | - Mario Paolo Colombo
- Department of Research, Molecular Immunology Unit, Fondazione IRCCS Istituto Nazionale Dei Tumori, via Amadeo 42, 20133, Milano, Italy.
| |
Collapse
|
129
|
Liu C, Piao J, Shang Z. Hyperprogressive disease after radiotherapy combined with anti-PD-1 therapy in renal cell carcinoma: a case report and review of the literature. BMC Urol 2021; 21:42. [PMID: 33743675 PMCID: PMC7981866 DOI: 10.1186/s12894-021-00813-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/16/2021] [Indexed: 01/10/2023] Open
Abstract
Background Studies have shown that immune checkpoint inhibitors (ICIs) have limited efficacy and can even increase tumour burden in short time periods. This is usually called hyperprogressive disease (HPD). To date, there are few reports regarding HPD; fewer have analysed the relationship between HPD and radiotherapy combined with ICIs, and their conclusions are controversial. Case presentation A 42-year-old woman was diagnosed with stage IV renal clear cell carcinoma. The patient had previously received sorafenib and pazopanib as first- and second-line therapies, respectively. She received radiotherapy combined with nivolumab. Eighteen days after administration of the third dose of nivolumab, the patient’s general condition deteriorated; this was associated with immune-related adverse events. Computed tomography showed that the diameter of left lung metastases had sharply increased. A biopsy of the lung metastasis showed no infiltration of lymphocytes. The patient’s general condition worsened and she died of the disease on the 70th day after administration of the third dose of nivolumab. Conclusions This report describes the development of HPD following the administration of radiotherapy combined with ICIs in a case of advanced renal cell carcinoma. The case indicates that radiotherapy may show bidirectional regulation effects on anti-tumour immune response. If the immunosuppressive function of radiotherapy is dominant, combined with ICIs, it could result in HPD. Supplementary Information The online version contains supplementary material available at 10.1186/s12894-021-00813-8.
Collapse
Affiliation(s)
- Chao Liu
- Department of Oncology, Zhuozhou Hospital, No.129, Fanyang Road, Zhuozhou City, Hebei Province, China
| | - Jingjing Piao
- College of Nursing, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province, China
| | - Zhiyang Shang
- Department of Oncology, Zhuozhou Hospital, No.129, Fanyang Road, Zhuozhou City, Hebei Province, China.
| |
Collapse
|
130
|
Prelaj A, Pircher CC, Massa G, Martelli V, Corrao G, Lo Russo G, Proto C, Ferrara R, Galli G, De Toma A, Genova C, Jereczek-Fossa BA, de Braud F, Garassino MC, Rebuzzi SE. Beyond First-Line Immunotherapy: Potential Therapeutic Strategies Based on Different Pattern Progressions: Oligo and Systemic Progression. Cancers (Basel) 2021; 13:1300. [PMID: 33803958 PMCID: PMC7999258 DOI: 10.3390/cancers13061300] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 02/07/2023] Open
Abstract
First-line immune-checkpoint inhibitor (ICI)-based therapy has deeply changed the treatment landscape and prognosis in advanced non-small cell lung cancer (aNSCLC) patients with no targetable alterations. Nonetheless, a percentage of patients progressed on ICI as monotherapy or combinations. Open questions remain on patients' selection, the identification of biomarkers of primary resistance to immunotherapy and the treatment strategies to overcome secondary resistance to first-line immunotherapy. Local ablative approaches are the main therapeutic strategies in oligoprogressive disease, and their role is emerging in patients treated with immunotherapy. Many therapeutic strategies can be adapted in aNSCLC patients with systemic progression to personalize the treatment approach according to re-characterization of the tumors, previous ICI response, and type of progression. This review's aim is to highlight and discuss the current and potential therapeutic approaches beyond first-line ICI-based therapy in aNSCLC patients based on the pattern of disease progression (oligoprogression versus systemic progression).
Collapse
Affiliation(s)
- Arsela Prelaj
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, via Giacomo Venezian 1, 20133 Milan, Italy; (C.C.P.); (G.M.); (G.L.R.); (C.P.); (R.F.); (G.G.); (A.D.T.); (F.d.B.); (M.C.G.)
- Department of Electronics, Information, and Bioengineering, Polytechnic University of Milan, Piazza Leonardo Da Vinci 32, 20133 Milan, Italy
| | - Chiara Carlotta Pircher
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, via Giacomo Venezian 1, 20133 Milan, Italy; (C.C.P.); (G.M.); (G.L.R.); (C.P.); (R.F.); (G.G.); (A.D.T.); (F.d.B.); (M.C.G.)
| | - Giacomo Massa
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, via Giacomo Venezian 1, 20133 Milan, Italy; (C.C.P.); (G.M.); (G.L.R.); (C.P.); (R.F.); (G.G.); (A.D.T.); (F.d.B.); (M.C.G.)
| | - Valentino Martelli
- Oncologia Medica 1, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (V.M.); or (S.E.R.)
| | - Giulia Corrao
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy; (G.C.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, via Festa del Perdono, 7, 20122 Milan, Italy
| | - Giuseppe Lo Russo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, via Giacomo Venezian 1, 20133 Milan, Italy; (C.C.P.); (G.M.); (G.L.R.); (C.P.); (R.F.); (G.G.); (A.D.T.); (F.d.B.); (M.C.G.)
| | - Claudia Proto
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, via Giacomo Venezian 1, 20133 Milan, Italy; (C.C.P.); (G.M.); (G.L.R.); (C.P.); (R.F.); (G.G.); (A.D.T.); (F.d.B.); (M.C.G.)
| | - Roberto Ferrara
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, via Giacomo Venezian 1, 20133 Milan, Italy; (C.C.P.); (G.M.); (G.L.R.); (C.P.); (R.F.); (G.G.); (A.D.T.); (F.d.B.); (M.C.G.)
| | - Giulia Galli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, via Giacomo Venezian 1, 20133 Milan, Italy; (C.C.P.); (G.M.); (G.L.R.); (C.P.); (R.F.); (G.G.); (A.D.T.); (F.d.B.); (M.C.G.)
| | - Alessandro De Toma
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, via Giacomo Venezian 1, 20133 Milan, Italy; (C.C.P.); (G.M.); (G.L.R.); (C.P.); (R.F.); (G.G.); (A.D.T.); (F.d.B.); (M.C.G.)
| | - Carlo Genova
- UO Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy;
- Dipartimento di Medicina Interna e Specialità Mediche (DiMI), Università degli Studi di Genova, Viale Benedetto XV 6, 16132 Genoa, Italy
| | - Barbara Alicja Jereczek-Fossa
- Division of Radiation Oncology, IEO, European Institute of Oncology IRCCS, via Ripamonti 435, 20141 Milan, Italy; (G.C.); (B.A.J.-F.)
- Department of Oncology and Hemato-Oncology, University of Milan, via Festa del Perdono, 7, 20122 Milan, Italy
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, via Giacomo Venezian 1, 20133 Milan, Italy; (C.C.P.); (G.M.); (G.L.R.); (C.P.); (R.F.); (G.G.); (A.D.T.); (F.d.B.); (M.C.G.)
| | - Marina Chiara Garassino
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale Tumori, via Giacomo Venezian 1, 20133 Milan, Italy; (C.C.P.); (G.M.); (G.L.R.); (C.P.); (R.F.); (G.G.); (A.D.T.); (F.d.B.); (M.C.G.)
| | - Sara Elena Rebuzzi
- Oncologia Medica 1, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genova, Italy; (V.M.); or (S.E.R.)
- Dipartimento di Medicina Interna e Specialità Mediche (DiMI), Università degli Studi di Genova, Viale Benedetto XV 6, 16132 Genoa, Italy
| |
Collapse
|
131
|
Yue Q, Xu Y, Deng X, Wang S, Qiu J, Qian B, Zhang Y. Circ-PITX1 Promotes the Progression of Non-Small Cell Lung Cancer Through Regulating the miR-1248/CCND2 Axis. Onco Targets Ther 2021; 14:1807-1819. [PMID: 33727831 PMCID: PMC7955706 DOI: 10.2147/ott.s286820] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/28/2021] [Indexed: 12/15/2022] Open
Abstract
Background Circular RNA (circRNA) is a key regulator of cancer, and it has been proved to be involved in the regulation of cancer progression including non-small cell lung cancer (NSCLC). Circ-PITX1 was found to be a significantly upregulated circRNA in NSCLC, and its role and potential mechanism in NSCLC progression deserve further investigation. Methods The expression levels of circ-PITX1, microRNA (miR)-1248 and cyclin D2 (CCND2) were examined by quantitative real-time PCR (qRT-PCR). Cell proliferation, apoptosis, cell cycle process, migration and invasion were determined using cell counting kit 8 (CCK8) assay, colony formation assay, flow cytometry, wound healing assay and transwell assay. Xenograft models were built to explore the role of circ-PITX1 in NSCLC tumor growth in vivo. The glycolysis and glutamine metabolism of cells were assessed by detecting the consumptions of glucose and glutamine, cell extracellular acidification rate (ECAR), and the productions of lactate, α-ketoglutaric acid (α-KG) and ATP. The protein levels of hexokinase 2 (HK-2), glutaminase 1 (GLS1) and CCND2 were tested by Western blot (WB) analysis. Dual-luciferase reporter assay and RIP assay were employed to verify the interaction between miR-1248 and circ-PITX1 or CCND2. Results Circ-PITX1 was upregulated in NSCLC and its silencing could inhibit the proliferation, migration, invasion, cell cycle process, glycolysis, glutamine metabolism, and promote the apoptosis of NSCLC cells in vitro, as well as reduced tumor growth in vivo. In the terms of mechanism, we found that circ-PITX1 could act as a sponge of miR-1248, and miR-1248 could target CCND2. In addition, miR-1248 inhibitor reversed the inhibitory effect of circ-PITX1 knockdown on NSCLC progression. Similarly, CCND2 overexpression also reversed the suppressive effect of miR-1248 on NSCLC progression. Moreover, circ-PITX1 positively regulated CCND2 expression by sponging miR-1248. Conclusion Circ-PITX1 served as a sponge of miR-1248 to promote NSCLC progression by upregulating CCND2.
Collapse
Affiliation(s)
- Qianyu Yue
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, 650032, Yunnan, People's Republic of China
| | - Yanyan Xu
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, 650032, Yunnan, People's Republic of China
| | - Xiaoli Deng
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, 650032, Yunnan, People's Republic of China
| | - Shenglan Wang
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, 650032, Yunnan, People's Republic of China
| | - Jingman Qiu
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, 650032, Yunnan, People's Republic of China
| | - Baojiang Qian
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, 650032, Yunnan, People's Republic of China
| | - Yunhui Zhang
- Department of Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, 650032, Yunnan, People's Republic of China
| |
Collapse
|
132
|
Park HJ, Kim KW, Won SE, Yoon S, Chae YK, Tirumani SH, Ramaiya NH. Definition, Incidence, and Challenges for Assessment of Hyperprogressive Disease During Cancer Treatment With Immune Checkpoint Inhibitors: A Systematic Review and Meta-analysis. JAMA Netw Open 2021; 4:e211136. [PMID: 33760090 PMCID: PMC7991969 DOI: 10.1001/jamanetworkopen.2021.1136] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
IMPORTANCE Hyperprogressive disease (HPD) is a recognized pattern of rapid tumor progression during immune checkpoint inhibitor (ICI) treatment. Definitions of HPD have not been standardized, posing the risk of capturing different tumoral behaviors. OBJECTIVES To provide a systematic summary of definitions and the incidence of HPD in patients undergoing ICI treatment and discuss the challenges of current assessment of HPD. DATA SOURCES Articles that evaluated HPD published before March 3, 2020, were identified from MEDLINE and EMBASE. STUDY SELECTION Clinical trials and observational studies providing the incidence and definition of HPD from patients with cancer treated with ICIs. DATA EXTRACTION AND SYNTHESIS Factors included in the analysis comprised authors, year of publication, cancer type, ICI type, number of previous treatment lines, definition of HPD, time frame used to assess HPD, number of patients with HPD, onset of HPD, and prognosis of patients with HPD. Quantitative and qualitative syntheses for the incidence of HPD were performed. MAIN OUTCOMES AND MEASURES Definitions of HPD were categorized and the range of incidence of HPD was evaluated. Subgroup analysis on the incidence of HPD according to the category was performed and the challenges associated with current HPD assessment were evaluated. RESULTS Twenty-four studies with 3109 patients were analyzed. The incidence of HPD varied from 5.9% to 43.1%. The definitions were divided into 4 categories based on the calculation of tumor growth acceleration: tumor growth rate ratio (pooled incidence of HPD, 9.4%; 95% CI, 6.9%-12.0%), tumor growth kinetics ratio (pooled incidence, 15.8%; 95% CI, 8.0%-23.7%), early tumor burden increase (pooled incidence, 20.6%; 95% CI, 9.3%-31.8%), and combinations of the above (pooled incidence, 12.4%; 95% CI, 7.3%-17.5%). Hyperprogressive disease could be overestimated or underestimated if the assessment was limited to tumor growth rate or tumor growth kinetics ratio, target lesions, or response evaluation criteria in solid tumors (RECIST)-defined progressors, or if the assessment time frame conformed to RECIST. Study results on clinical outcome were heterogeneous on discriminating patients with HPD from those with natural progressive disease. CONCLUSIONS AND RELEVANCE Definitions of HPD appear to be diverse, with the incidence of HPD varying from 5.9% to 43.1% across studies examined in this meta-analysis. Varying incidence and definitions of HPD indicate the need for establishing its uniform and clinically relevant criteria based on currently available evidence.
Collapse
Affiliation(s)
- Hyo Jung Park
- Asan Image Metrics, Asan Medical Center, Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Kyung Won Kim
- Asan Image Metrics, Asan Medical Center, Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang Eun Won
- Asan Image Metrics, Asan Medical Center, Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Shinkyo Yoon
- Asan Medical Center, Department of Oncology, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Young Kwang Chae
- Feinberg School of Medicine, Robert H. Lurie Comprehensive Cancer Center, Department of Medicine, Northwestern University, Chicago, Illinois
| | - Sree Harsha Tirumani
- University Hospitals Cleveland Medical Center, Department of Radiology, Case Western Reserve University, Cleveland, Ohio
| | - Nikhil H. Ramaiya
- University Hospitals Cleveland Medical Center, Department of Radiology, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
133
|
Affiliation(s)
- Kartik Sehgal
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
134
|
Makuku R, Khalili N, Razi S, Keshavarz-Fathi M, Rezaei N. Current and Future Perspectives of PD-1/PDL-1 Blockade in Cancer Immunotherapy. J Immunol Res 2021; 2021:6661406. [PMID: 33681388 PMCID: PMC7925068 DOI: 10.1155/2021/6661406] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 01/23/2021] [Accepted: 02/10/2021] [Indexed: 12/18/2022] Open
Abstract
Cancer immunotherapy, which reactivates weakened immune cells of cancer patients, has yielded great success in recent years. Among immunotherapeutic agents, immune checkpoint inhibitors have been of particular interest and have gained approval by the FDA for treatment of cancers. Immune checkpoint blockade through targeting programmed cell death protein-1 (PD-1) has demonstrated promising antitumor effects in cancer immunotherapy of many different solid and hematologic malignancies. However, despite promising results, a favorable response is observed only in a fraction of patients, and there is still lack of a single therapy modality with curative ability. In this paper, we review the current and future perspectives of PD-1/L1 blockade in cancer immunotherapy, with a particular focus on predictive biomarkers of response to therapy. We also discuss the adverse events associated with PD-1/L1/2 inhibitors, ranging from severe life-threatening conditions such as autoimmune myocarditis to mild and moderate reactions such as skin rashes, and explore the potential strategies for improving the efficacy of immunotherapy with PD-1/L1 checkpoint inhibitors.
Collapse
Affiliation(s)
- Rangarirai Makuku
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Neda Khalili
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Student Research Committee, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Keshavarz-Fathi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Sheffield, UK
| |
Collapse
|
135
|
Qu J, Mei Q, Liu L, Cheng T, Wang P, Chen L, Zhou J. The progress and challenge of anti-PD-1/PD-L1 immunotherapy in treating non-small cell lung cancer. Ther Adv Med Oncol 2021; 13:1758835921992968. [PMID: 33643442 PMCID: PMC7890731 DOI: 10.1177/1758835921992968] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
The use of programmed cell-death protein 1 (PD-1)/programmed cell-death ligand 1 (PD-L1) inhibitors is the standard therapy for the first-line or second-line treatment of patients with non-small-cell lung cancer (NSCLC). In contrast to current traditional treatments such as chemotherapy or radiotherapy, anti-PD-1 and anti-PD-L1 treatments can directly attenuate tumour-mediated exhaustion and effectively modulate the host anti-tumour immune response in vivo. In addition, compared with traditional therapy, PD-1/PD-L1 inhibitor monotherapy can significantly prolong survival without obvious side effects in the treatment of advanced NSCLC. Ideally, several biomarkers could be used to monitor the safety and effectiveness of anti-PD-1 and anti-PD-L1 treatments; however, the current lack of optimal prognostic markers remains a widespread limitation and challenge for further clinical applications, as does the possibility of immune-related adverse events and drug resistance. In this review, we aimed to summarise the latest progress in anti-PD-1/anti-PD-L1 treatment of advanced NSCLC, worldwide, including in China. An exploration of underlying biomarker identification and future challenges will be discussed in this article to facilitate translational studies in cancer immunotherapy.
Collapse
Affiliation(s)
- Jingjing Qu
- Department of Respiratory Disease, Thoracic Disease Centre, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Quanhui Mei
- Intensive Care Unit, The First People’s Hospital of Changde City, Changde, Hunan, PR China
| | - Li Liu
- Lung Cancer and Gastroenterology Department, Hunan Cancer Hospital, Affiliated Tumour Hospital of Xiangya Medical School of Central South University, Changsha, Hunan, PR China
| | - Tianli Cheng
- Thoracic Medicine Department 1, Hunan Cancer Hospital, Affiliated Tumour Hospital of Xiangya Medical School of Central South University, Changsha, Hunan, PR China
| | - Peng Wang
- Ningxia Key Laboratory of Cerebrocranial Diseases, School of Basic Medical Science, Ningxia Medical University, Yinchuan, Ningxia, PR China
| | - Lijun Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Centre for Infectious Diseases, Collaborative Innovation Centre for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003, PR China
| | - Jianying Zhou
- Department of Respiratory Disease, Thoracic Disease Centre, The First Affiliated Hospital, College of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang 310003, PR China
| |
Collapse
|
136
|
Sato Y, Morimoto T, Hara S, Nagata K, Hosoya K, Nakagawa A, Tachikawa R, Tomii K. Dissociated response and clinical benefit in patients treated with nivolumab monotherapy. Invest New Drugs 2021; 39:1170-1178. [PMID: 33566254 DOI: 10.1007/s10637-021-01077-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/28/2021] [Indexed: 12/25/2022]
Abstract
Immune checkpoint inhibitors (ICIs) are effective for previously treated patients with advanced non-small cell lung cancer (NSCLC). However, an unconventional response pattern is sometimes encountered. A dissociated response (DR), characterized by some lesions shrinking and others growing, has been recognized with ICI treatment. In this study, we examined the characteristics and treatment outcomes of DR in previously treated NSCLC patients, receiving nivolumab monotherapy. We conducted a retrospective cohort study of previously treated patients with advanced NSCLC who received nivolumab. We assessed the tumor response of each organ using the Response Evaluation Criteria in Solid Tumors (RECIST) criteria at the first radiologic evaluation. We investigated treatment outcome and compared overall survival using the Kaplan-Meier Method and log-rank tests. Further, we conducted the same analysis in patients who had previously received chemotherapy or tyrosine kinase inhibitor therapy in our hospital. Between April 2016 and September 2018, 107 patients who received nivolumab fulfilled the inclusion criteria. Of them, 5 (5%) patients showed a DR. There were no specific differences in characteristics between DR and non-DR cases. Patients showing DR had significantly longer overall survival than those showing concordant progressive disease (46.9 vs. 8.2 months, p = 0.038). The frequencies of DR in the ICI, chemotherapy, and tyrosine kinase inhibitor-treated cohorts were 5%, 1%, and 4%, respectively. DR was uncommon, but this presented a distinctive pattern of nivolumab response. Some patients might benefit from continuing nivolumab therapy and may achieve a longer overall survival.
Collapse
Affiliation(s)
- Yuki Sato
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, 2-1-1 Minatojima-Minamimachi, Chuo-ku, Kobe, 650-0047, Japan.
| | - Takeshi Morimoto
- Department of Clinical Research Center, Kobe City Medical Center General Hospital, Kobe, 650-0047, Japan.,Department of Clinical Epidemiology, Hyogo College of Medicine, Nishinomiya, 663-8501, Japan
| | - Shigeo Hara
- Department of Clinical Pathology, Kobe City Medical Center General Hospital, Kobe, 650-0047, Japan
| | - Kazuma Nagata
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, 2-1-1 Minatojima-Minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Kazutaka Hosoya
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, 2-1-1 Minatojima-Minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Atsushi Nakagawa
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, 2-1-1 Minatojima-Minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Ryo Tachikawa
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, 2-1-1 Minatojima-Minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| | - Keisuke Tomii
- Department of Respiratory Medicine, Kobe City Medical Center General Hospital, 2-1-1 Minatojima-Minamimachi, Chuo-ku, Kobe, 650-0047, Japan
| |
Collapse
|
137
|
Barham W, Guo R, Park SS, Herrmann J, Dong H, Yan Y. Case Report: Simultaneous Hyperprogression and Fulminant Myocarditis in a Patient With Advanced Melanoma Following Treatment With Immune Checkpoint Inhibitor Therapy. Front Immunol 2021; 11:561083. [PMID: 33603731 PMCID: PMC7884751 DOI: 10.3389/fimmu.2020.561083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 12/17/2020] [Indexed: 01/05/2023] Open
Abstract
We report here a patient with stage IV mucosal melanoma treated with dual immune checkpoint inhibitor (ICI) therapy (Nivolumab/Ipilimumab) who experienced rapid disease progression and metastatic spread within three weeks of first infusion. Surprisingly, this patient also developed fulminant myocarditis within the same time frame. Immunohistochemical staining of the primary tumor and a metastatic omental lesion revealed robust CD8+ PD-1+ T cell infiltration after ICI treatment, as would be expected following immune activation. However, the CD8+ T cell infiltrate was largely negative for both Granzyme B and TIA-1, suggesting these T cells were not capable of effective tumor lysis. We discuss the possibility that heightened pro-inflammatory T cell activity (rather than tumor-directed cytolytic activity) was induced by anti-PD-1 and anti-CTLA-4, which could have provoked both rapid tumor resistance mechanisms and myocarditis. This case highlights the fact that the mere presence of tumor infiltrating lymphocytes (TILs) does not necessarily correlate to ICI response and that additional functional markers are necessary to differentiate between inflammatory and cytolytic CD8+ TILs.
Collapse
Affiliation(s)
- Whitney Barham
- Department of Immunology, Mayo Clinic, Rochester, MN, United States.,Medical Scientist Training Program, Mayo Clinic, Rochester, MN, United States
| | - Ruifeng Guo
- Dermatopathology Section, Division of Anatomic Pathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Sean S Park
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN, United States
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, United States
| | - Haidong Dong
- Department of Immunology, Mayo Clinic, Rochester, MN, United States.,Department of Urology, Mayo Clinic, Rochester, MN, United States
| | - Yiyi Yan
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
138
|
Kim CG, Kim C, Yoon SE, Kim KH, Choi SJ, Kang B, Kim HR, Park SH, Shin EC, Kim YY, Kim DJ, Chung HC, Chon HJ, Choi HJ, Lim HY. Hyperprogressive disease during PD-1 blockade in patients with advanced hepatocellular carcinoma. J Hepatol 2021; 74:350-359. [PMID: 32810553 DOI: 10.1016/j.jhep.2020.08.010] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/03/2020] [Accepted: 08/08/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Programmed cell death-1 (PD-1) inhibitor treatment can cause hyperprogressive disease (HPD), but the incidence, outcome, and predictive factors of HPD are unknown in patients with hepatocellular carcinoma (HCC). Herein, we assessed the existence and factors predictive of HPD in patients with advanced HCC treated with nivolumab. METHODS We enrolled 189 patients with advanced HCC treated with nivolumab. Occurrence of HPD was investigated using tumour growth dynamics based on tumour growth kinetics (TGK) and tumour growth rate (TGR) before and after treatment, or time to treatment failure. We additionally analysed patients treated with regorafenib (n = 95) or best supportive care (BSC)/placebo (n = 103) after progression on sorafenib to compare tumour growth dynamics. RESULTS Flare-up of tumour growth was observed in a fraction of patients upon PD-1 blockade, indicating the occurrence of HPD. Based on distinct patterns of disease progression exclusively observed in the nivolumab-treated cohort, but not in the regorafenib- or BSC/placebo-treated cohorts, 4-fold increases in TGK and TGR ratios as well as a 40% increase in TGR were the cut-off values used to define HPD; 12.7% of the patients (24/189) treated with nivolumab met all these criteria. Patients with HPD had worse progression-free survival (hazard ratio [HR] 2.194; 95% CI 1.214-3.964) and overall survival (HR 2.238; 95% CI 1.233-4.062) compared to patients with progressive disease without HPD. More than 90% of patients with HPD missed the opportunity for subsequent treatment because of rapid clinical deterioration. An elevated neutrophil-to-lymphocyte ratio (>4.125) was associated with HPD and an inferior survival rate. CONCLUSIONS HPD occurs in a fraction of patients with HCC who receive PD-1 inhibitor treatment. Analyses of the baseline immune profile and on-treatment tumour growth dynamics could enable optimal patient selection and earlier identification of HPD. LAY SUMMARY Hyperprogressive disease is an unexpected response pattern observed in patients treated with an immune checkpoint inhibitor. This study revealed that hyperprogressive disease occurs in a fraction of patients with advanced hepatocellular carcinoma treated with an anti-PD-1 antibody, providing evidence to encourage careful monitoring of patients to prevent clinical deterioration induced by PD-1 blockade.
Collapse
Affiliation(s)
- Chang Gon Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea; Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Chan Kim
- Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Sang Eun Yoon
- Division of Hemato-Oncology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyung Hwan Kim
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea; Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Seong Jin Choi
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Beodeul Kang
- Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Hye Ryun Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Su-Hyung Park
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Yeun-Yoon Kim
- Department of Radiology, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Dae Jung Kim
- Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Korea
| | - Hyun Cheol Chung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea
| | - Hong Jae Chon
- Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, Korea.
| | - Hye Jin Choi
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Korea.
| | - Ho Yeong Lim
- Division of Hemato-Oncology, Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
139
|
Zhou F, Qiao M, Zhou C. The cutting-edge progress of immune-checkpoint blockade in lung cancer. Cell Mol Immunol 2021; 18:279-293. [PMID: 33177696 PMCID: PMC8027847 DOI: 10.1038/s41423-020-00577-5] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/15/2020] [Indexed: 12/24/2022] Open
Abstract
Great advances in immune checkpoint blockade have resulted in a paradigm shift in patients with lung cancer. Immune-checkpoint inhibitor (ICI) treatment, either as monotherapy or combination therapy, has been established as the standard of care for patients with locally advanced/metastatic non-small cell lung cancer without EGFR/ALK alterations or extensive-stage small cell lung cancer. An increasing number of clinical trials are also ongoing to further investigate the role of ICIs in patients with early-stage lung cancer as neoadjuvant or adjuvant therapy. Although PD-L1 expression and tumor mutational burden have been widely studied for patient selection, both of these biomarkers are imperfect. Due to the complex cancer-immune interactions among tumor cells, the tumor microenvironment and host immunity, collaborative efforts are needed to establish a multidimensional immunogram to integrate complementary predictive biomarkers for personalized immunotherapy. Furthermore, as a result of the wide use of ICIs, managing acquired resistance to ICI treatment remains an inevitable challenge. A deeper understanding of the underlying biological mechanisms of acquired resistance to ICIs is helpful to overcome these obstacles. In this review, we describe the cutting-edge progress made in patients with lung cancer, the optimal duration of ICI treatment, ICIs in some special populations, the unique response patterns during ICI treatment, the emerging predictive biomarkers, and our understanding of primary and acquired resistance mechanisms to ICI treatment.
Collapse
Affiliation(s)
- Fei Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Meng Qiao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Thoracic Cancer Institute, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
140
|
Hyperprogression in hepatocellular carcinoma: Illusion or reality? J Hepatol 2021; 74:269-271. [PMID: 33109354 DOI: 10.1016/j.jhep.2020.09.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 09/17/2020] [Indexed: 12/29/2022]
|
141
|
Kas B, Talbot H, Ferrara R, Richard C, Lamarque JP, Pitre-Champagnat S, Planchard D, Balleyguier C, Besse B, Mezquita L, Lassau N, Caramella C. Clarification of Definitions of Hyperprogressive Disease During Immunotherapy for Non-Small Cell Lung Cancer. JAMA Oncol 2021; 6:1039-1046. [PMID: 32525513 DOI: 10.1001/jamaoncol.2020.1634] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Importance Hyperprogressive disease (HPD) is an aggressive pattern of progression reported for patients treated with programmed cell death 1 (PD-1)/programmed cell death 1 ligand (PD-L1) inhibitors as a single agent in several studies. However, the use of different definitions of HPD introduces the risk of describing different tumoral behaviors. Objective To assess the accuracy of each HPD definition to identify the frequency of HPD and the association with poorer outcomes of immune-checkpoint inhibitor (ICI) treatment in patients with advanced non-small cell lung cancer (NSCLC) and to provide an optimized and homogenized definition based on all previous criteria for identifying HPD. Design, Setting, and Participants This retrospective cohort study included 406 patients with advanced NSCLC treated with PD-1/PD-L1 inhibitors from November 1, 2012, to April 5, 2017, in 8 French institutions. Measurable lesions were defined using the Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 criteria on at least 2 computed tomographic scans before the initiation of ICI therapy and 1 computed tomographic scan during treatment. Data were analyzed from November 1, 2012, to August 1, 2019. Exposures Advanced NSCLC and treatment with PD-1/PD-L1 inhibitors. Main Outcomes and Measures Association of the definition with the related incidence and the HPD subset constitution and the association between each HPD definition and overall survival. All dynamic indexes used in the previous proposed definitions, such as the tumor growth rate (TGR) or tumor growth kinetics (TGK), were calculated before and during treatment. Results Among the 406 patients with NSCLC included in the analysis (259 male [63.8%]; median age at start of ICI treatment, 64 [range, 30-91] years), the different definitions resulted in incidences of the HPD phenomenon varying from 5.4% (n = 22; definition based on a progression pace >2-fold and a time to treatment failure of <2 months) to 18.5% (n = 75; definition based on the TGR ratio). The concordance between these different definitions (using the Jaccard similarity index) varied from 33.3% to 69.3%. For every definition, HPD was associated with poorer survival (range of median overall survival, 3.4 [95% CI, 1.9-8.4] to 6.0 [95% CI, 3.7-9.4] months). The difference between TGR before and during therapy (ΔTGR) was the most correlated with poor overall survival with an initial plateau for a larger number of patients and a slower increase, and it had the highest ability to distinguish patients with HPD from those with progressive disease not classified as HPD. In addition, an optimal threshold of ΔTGR of greater than 100 was identified for this distinction. Conclusions and Relevance The findings of this retrospective cohort study of patients with NSCLC suggest that the previous 5 definitions of HPD were not associated with the same tumor behavior. A new definition, based on ΔTGR of greater than 100, appeared to be associated with the characteristics expected with HPD (increase of the tumor kinetics and poor survival). Additional studies on larger groups of patients are necessary to confirm the accuracy and validate this proposed definition.
Collapse
Affiliation(s)
- Baptiste Kas
- UMR (Unité Mixte de Recherche) 1281, Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Commissariat à l'énergie Atomique et Aux Énergies Alternatives, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Villejuif, France
| | - Hugues Talbot
- Center for Visual Computing, CentraleSupelec, Inria, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Roberto Ferrara
- Department of Medical Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Istituto Nazionale dei Tumori, Milan, Italy
| | - Colombe Richard
- UMR (Unité Mixte de Recherche) 1281, Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Commissariat à l'énergie Atomique et Aux Énergies Alternatives, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Villejuif, France
| | - Jean-Philippe Lamarque
- UMR (Unité Mixte de Recherche) 1281, Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Commissariat à l'énergie Atomique et Aux Énergies Alternatives, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Villejuif, France
| | - Stéphanie Pitre-Champagnat
- UMR (Unité Mixte de Recherche) 1281, Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Commissariat à l'énergie Atomique et Aux Énergies Alternatives, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Villejuif, France
| | - David Planchard
- Cancer Medicine Department, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Corinne Balleyguier
- UMR (Unité Mixte de Recherche) 1281, Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Commissariat à l'énergie Atomique et Aux Énergies Alternatives, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Villejuif, France.,Radiology Department, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Benjamin Besse
- Cancer Medicine Department, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Laura Mezquita
- Cancer Medicine Department, Gustave Roussy, Université Paris-Saclay, Villejuif, France.,Medical Oncology Department, Hospital Clínic, Barcelona, Spain.,Translational Genomics and Targeted Therapeutics in Solid Tumours, August Pi i Sunyer Biomedical Research Institute, Barcelona, Spain
| | - Nathalie Lassau
- UMR (Unité Mixte de Recherche) 1281, Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Commissariat à l'énergie Atomique et Aux Énergies Alternatives, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Villejuif, France.,Radiology Department, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Caroline Caramella
- UMR (Unité Mixte de Recherche) 1281, Université Paris-Saclay, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Commissariat à l'énergie Atomique et Aux Énergies Alternatives, Laboratoire d'Imagerie Biomédicale Multimodale Paris-Saclay, Villejuif, France.,Radiology Department, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
142
|
Angelicola S, Ruzzi F, Landuzzi L, Scalambra L, Gelsomino F, Ardizzoni A, Nanni P, Lollini PL, Palladini A. IFN-γ and CD38 in Hyperprogressive Cancer Development. Cancers (Basel) 2021; 13:309. [PMID: 33467713 PMCID: PMC7830527 DOI: 10.3390/cancers13020309] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 01/11/2021] [Accepted: 01/13/2021] [Indexed: 12/21/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) improve the survival of patients with multiple types of cancer. However, low response rates and atypical responses limit their success in clinical applications. The paradoxical acceleration of tumor growth after treatment, defined as hyperprogressive disease (HPD), is the most difficult problem facing clinicians and patients alike. The mechanisms that underlie hyperprogression (HP) are still unclear and controversial, although different factors are associated with the phenomenon. In this review, we propose two factors that have not yet been demonstrated to be directly associated with HP, but upon which it is important to focus attention. IFN-γ is a key cytokine in antitumor response and its levels increase during ICI therapy, whereas CD38 is an alternative immune checkpoint that is involved in immunosuppressive responses. As both factors are associated with resistance to ICI therapy, we have discussed their possible involvement in HPD with the conclusion that IFN-γ may contribute to HP onset through the activation of the inflammasome pathway, immunosuppressive enzyme IDO1 and activation-induced cell death (AICD) in effector T cells, while the role of CD38 in HP may be associated with the activation of adenosine receptors, hypoxia pathways and AICD-dependent T-cell depletion.
Collapse
Affiliation(s)
- Stefania Angelicola
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy; (S.A.); (F.R.); (L.S.); (A.P.)
| | - Francesca Ruzzi
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy; (S.A.); (F.R.); (L.S.); (A.P.)
| | - Lorena Landuzzi
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Laura Scalambra
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy; (S.A.); (F.R.); (L.S.); (A.P.)
| | - Francesco Gelsomino
- Divisione di Oncologia Medica, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.G.); (A.A.)
| | - Andrea Ardizzoni
- Divisione di Oncologia Medica, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (F.G.); (A.A.)
| | - Patrizia Nanni
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy; (S.A.); (F.R.); (L.S.); (A.P.)
| | - Pier-Luigi Lollini
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy; (S.A.); (F.R.); (L.S.); (A.P.)
| | - Arianna Palladini
- Laboratory of Immunology and Biology of Metastasis, Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126 Bologna, Italy; (S.A.); (F.R.); (L.S.); (A.P.)
| |
Collapse
|
143
|
Volk V, Theobald SJ, Danisch S, Khailaie S, Kalbarczyk M, Schneider A, Bialek-Waldmann J, Krönke N, Deng Y, Eiz-Vesper B, Dragon AC, von Kaisenberg C, Lienenklaus S, Bleich A, Keck J, Meyer-Hermann M, Klawonn F, Hammerschmidt W, Delecluse HJ, Münz C, Feuerhake F, Stripecke R. PD-1 Blockade Aggravates Epstein-Barr Virus + Post-Transplant Lymphoproliferative Disorder in Humanized Mice Resulting in Central Nervous System Involvement and CD4 + T Cell Dysregulations. Front Oncol 2021; 10:614876. [PMID: 33511078 PMCID: PMC7837057 DOI: 10.3389/fonc.2020.614876] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/16/2020] [Indexed: 12/17/2022] Open
Abstract
Post-transplant lymphoproliferative disorder (PTLD) is one of the most common malignancies after solid organ or allogeneic stem cell transplantation. Most PTLD cases are B cell neoplasias carrying Epstein-Barr virus (EBV). A therapeutic approach is reduction of immunosuppression to allow T cells to develop and combat EBV. If this is not effective, approaches include immunotherapies such as monoclonal antibodies targeting CD20 and adoptive T cells. Immune checkpoint inhibition (ICI) to treat EBV+ PTLD was not established clinically due to the risks of organ rejection and graft-versus-host disease. Previously, blockade of the programmed death receptor (PD)-1 by a monoclonal antibody (mAb) during ex vivo infection of mononuclear cells with the EBV/M81+ strain showed lower xenografted lymphoma development in mice. Subsequently, fully humanized mice infected with the EBV/B95-8 strain and treated in vivo with a PD-1 blocking mAb showed aggravation of PTLD and lymphoma development. Here, we evaluated vis-a-vis in fully humanized mice after EBV/B95-8 or EBV/M81 infections the effects of a clinically used PD-1 blocker. Fifteen to 17 weeks after human CD34+ stem cell transplantation, Nod.Rag.Gamma mice were infected with two types of EBV laboratory strains expressing firefly luciferase. Dynamic optical imaging analyses showed systemic EBV infections and this triggered vigorous human CD8+ T cell expansion. Pembrolizumab administered from 2 to 5 weeks post-infections significantly aggravated EBV systemic spread and, for the M81 model, significantly increased the mortality of mice. ICI promoted Ki67+CD30+CD20+EBER+PD-L1+ PTLD with central nervous system (CNS) involvement, mirroring EBV+ CNS PTLD in humans. PD-1 blockade was associated with lower frequencies of circulating T cells in blood and with a profound collapse of CD4+ T cells in lymphatic tissues. Mice treated with pembrolizumab showed an escalation of exhausted T cells expressing TIM-3, and LAG-3 in tissues, higher levels of several human cytokines in plasma and high densities of FoxP3+ regulatory CD4+ and CD8+ T cells in the tumor microenvironment. We conclude that PD-1 blockade during acute EBV infections driving strong CD8+ T cell priming decompensates T cell development towards immunosuppression. Given the variety of preclinical models available, our models conferred a cautionary note indicating that PD-1 blockade aggravated the progression of EBV+ PTLD.
Collapse
Affiliation(s)
- Valery Volk
- Laboratory of Regenerative Immune Therapies Applied, REBIRTH - Research Center for Translational Regenerative Medicine, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany.,German Centre for Infection Research (DZIF), Partner site Hannover, Hannover, Germany.,Institute for Pathology, Hannover Medical School, Hannover, Germany
| | - Sebastian J Theobald
- Laboratory of Regenerative Immune Therapies Applied, REBIRTH - Research Center for Translational Regenerative Medicine, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany.,German Centre for Infection Research (DZIF), Partner site Hannover, Hannover, Germany
| | - Simon Danisch
- Laboratory of Regenerative Immune Therapies Applied, REBIRTH - Research Center for Translational Regenerative Medicine, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany.,German Centre for Infection Research (DZIF), Partner site Hannover, Hannover, Germany
| | - Sahamoddin Khailaie
- Department of Systems Immunology, Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Maja Kalbarczyk
- Laboratory of Regenerative Immune Therapies Applied, REBIRTH - Research Center for Translational Regenerative Medicine, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany.,German Centre for Infection Research (DZIF), Partner site Hannover, Hannover, Germany
| | - Andreas Schneider
- Laboratory of Regenerative Immune Therapies Applied, REBIRTH - Research Center for Translational Regenerative Medicine, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Julia Bialek-Waldmann
- Laboratory of Regenerative Immune Therapies Applied, REBIRTH - Research Center for Translational Regenerative Medicine, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Nicole Krönke
- Institute for Pathology, Hannover Medical School, Hannover, Germany
| | - Yun Deng
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Britta Eiz-Vesper
- Institute for Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Anna Christina Dragon
- Institute for Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Constantin von Kaisenberg
- Department of Obstetrics, Gynecology and Reproductive Medicine, Hannover Medical School, Hannover, Germany
| | - Stefan Lienenklaus
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Andre Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - James Keck
- The Jackson Laboratory, Sacramento, CA, United States
| | - Michael Meyer-Hermann
- Department of Systems Immunology, Braunschweig Integrated Centre of Systems Biology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Frank Klawonn
- Biostatistics Group, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute for Information Engineering, Ostfalia University, Wolfenbuettel, Germany
| | - Wolfgang Hammerschmidt
- Research Unit Gene Vectors, Helmholtz Zentrum München, German Research Center for Environmental Health and German Centre for Infection Research (DZIF), Partner site Munich, Munich, Germany
| | - Henri-Jacques Delecluse
- German Cancer Research Center (DKFZ), Institut National de la Santé et de la Recherche Médicale (INSERM) Unit U1074, Heidelberg, Germany
| | - Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland
| | - Friedrich Feuerhake
- Institute for Pathology, Hannover Medical School, Hannover, Germany.,Institute for Neuropathology, University Clinic Freiburg, Freiburg, Germany
| | - Renata Stripecke
- Laboratory of Regenerative Immune Therapies Applied, REBIRTH - Research Center for Translational Regenerative Medicine, Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany.,German Centre for Infection Research (DZIF), Partner site Hannover, Hannover, Germany
| |
Collapse
|
144
|
Kim SR, Chun SH, Kim JR, Kim SY, Seo JY, Jung CK, Gil BM, Kim JO, Ko YH, Woo IS, Shim BY, Hong SH, Kang JH. The implications of clinical risk factors, CAR index, and compositional changes of immune cells on hyperprogressive disease in non-small cell lung cancer patients receiving immunotherapy. BMC Cancer 2021; 21:19. [PMID: 33402127 PMCID: PMC7786505 DOI: 10.1186/s12885-020-07727-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/11/2020] [Indexed: 01/19/2023] Open
Abstract
Background Immune checkpoint blockades (ICBs) are characterized by a durable clinical response and better tolerability in patients with a variety of advanced solid tumors. However, we not infrequently encounter patients with hyperprogressive disease (HPD) exhibiting paradoxically accelerated tumor growth with poor clinical outcomes. This study aimed to investigate implications of clinical factors and immune cell composition on different tumor responses to immunotherapy in patients with non-small cell lung cancer (NSCLC). Methods This study evaluated 231 NSCLC patients receiving ICBs between January 2014 and May 2018. HPD was defined as a > 2-fold tumor growth kinetics ratio during ICB therapy and time-to-treatment failure of ≤2 months. We analyzed clinical data, imaging studies, periodic serologic indexes, and immune cell compositions in tumors and stromata using multiplex immunohistochemistry. Results Of 231 NSCLC patients, PR/CR and SD were observed in 50 (21.6%) and 79 (34.2%) patients, respectively and 26 (11.3%) patients met the criteria for HPD. Median overall survival in poor response groups (HPD and non-HPD PD) was extremely shorter than disease-controlled group (SD and PR/CR) (5.5 and 6.1 months vs. 16.2 and 18.3 months, respectively, P = 0.000). In multivariate analysis, HPD were significantly associated with heavy smoker (p = 0.0072), PD-L1 expression ≤1% (p = 0.0355), and number of metastatic site ≥3 (p = 0.0297). Among the serologic indexes including NLR, PLR, CAR, and LDH, only CAR had constantly significant correlations with HPD at the beginning of prior treatment and immunotherapy, and at the 1st tumor assessment. The number of CD4+ effector T cells and CD8+ cytotoxic T cells, and CD8+/PD-1+ tumor-infiltrating lymphocytes (TIL) tended to be smaller, especially in stromata of HPD group. More M2-type macrophages expressing CD14, CD68 and CD163 in the stromal area and markedly fewer CD56+ NK cells in the intratumoral area were observed in HPD group. Conclusions Our study suggests that not only clinical factors including heavy smoker, very low PD-L1 expression, multiple metastasis, and CAR index, but also fewer CD8+/PD-1+ TIL and more M2 macrophages in the tumor microenvironment are significantly associated with the occurrence of HPD in the patients with advanced/metastatic NSCLC receiving immunotherapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-020-07727-y.
Collapse
Affiliation(s)
- Seo Ree Kim
- Division of Medical Oncology, Department of Internal Medicine, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sang Hoon Chun
- Division of Medical Oncology, Department of Internal Medicine, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Joo Ri Kim
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Sang-Yeob Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jun Young Seo
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.,Department of Convergence Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Chan Kwon Jung
- Department of Hospital Pathology, College of Medicine, the Catholic University of Korea, Seoul, Republic of Korea
| | - Bo-Mi Gil
- Department of Radiology, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jeong-Oh Kim
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Yoon Ho Ko
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Medical Oncology, Department of Internal Medicine, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - In Sook Woo
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Medical Oncology, Department of Internal Medicine, Yeouido St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Byoung Yong Shim
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Division of Medical Oncology, Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sook-Hee Hong
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea.,Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.,Laboratory of Medical Oncology, Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jin Hyoung Kang
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222, Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea. .,Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea. .,Laboratory of Medical Oncology, Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
145
|
Zhang L, Wu L, Chen Q, Zhang B, Liu J, Liu S, Mo X, Li M, Chen Z, Chen L, You J, Jin Z, Chen X, Zhou Z, Zhang S. Predicting hyperprogressive disease in patients with advanced hepatocellular carcinoma treated with anti-programmed cell death 1 therapy. EClinicalMedicine 2021; 31:100673. [PMID: 33554079 PMCID: PMC7846667 DOI: 10.1016/j.eclinm.2020.100673] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/18/2020] [Accepted: 11/18/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Hyperprogressive disease (HPD) is a new progressive pattern in patients with advanced hepatocellular carcinoma (HCC) treated with programmed cell death 1 (PD-1) inhibitors. We aimed to investigate risk factors associated with HPD in advanced HCC patients undergoing anti-PD-1 therapy. METHODS A total of 69 patients treated with anti-PD-1 therapy between March 2017 and January 2020 were included. HPD was determined according to the time to treatment failure, tumour growth rate, and tumour growth rate ratio. Univariate and multivariate analyses were performed to identify clinical variables significantly associated with HPD. A risk model was constructed based on clinical variables with prognostic significance for HPD. FINDINGS Overall, 10 (14·49%) had HPD. Haemoglobin level, portal vein tumour thrombus, and Child-Pugh score were significantly associated with HPD. The risk model had an area under the curve of 0·931 (95% confidence interval, 0·844-1·000). Patients with HPD had a significantly shorter overall survival (OS) than that of the patients with non-HPD (p < 0·001). However, there was no significant difference in OS between PD (progressive disease) patients with and without HPD (p = 0·05). INTERPRETATION We identified three clinical variables as risk factors for HPD, providing an opportunity to aid the pre-treatment evaluation of the risk of HPD in patients treated with immunotherapy. FUNDING This study was funded by the National Natural Science Foundation of China (81571664, 81871323, and 81801665); National Natural Science Foundation of Guangdong Province (2018B030311024); Scientific Research General Project of Guangzhou Science Technology and Innovation Commission (201707010,328); and China Postdoctoral Science Foundation (2016M600145).
Collapse
Affiliation(s)
- Lu Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Lingeng Wu
- Department of Interventional Therapy, Cancer Centre, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Medical College Shantou University, Shantou, Guangdong, China
| | - Qiuying Chen
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Bin Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jing Liu
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Shuyi Liu
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xiaokai Mo
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Minmin Li
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zhuozhi Chen
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Luyan Chen
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Jingjing You
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Zhe Jin
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xudong Chen
- Minimally Invasive Interventional Treatment Centre, Shenzhen People's Hospital (The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University), Shenzhen, Guangdong, China
| | - Zejian Zhou
- Department of Interventional Therapy, Cancer Centre, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Shuixing Zhang
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
146
|
Definitions, outcomes, and management of hyperprogression in patients with non-small-cell lung cancer treated with immune checkpoint inhibitors. Lung Cancer 2020; 152:109-118. [PMID: 33385736 DOI: 10.1016/j.lungcan.2020.12.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 11/29/2020] [Accepted: 12/21/2020] [Indexed: 12/31/2022]
Abstract
BACKGROUND The advent of immune checkpoint inhibitors (ICI) has been a breakthrough in the care of patients with non-small-cell lung cancers (NSCLC). However, physicians are now facing a previously unidentified clinical situation called hyperprogression (HP), which presents as a fast and unexpected increase in tumor burden. HP's existence and specificity to ICIs remains controversial because a widely acknowledged definition is currently lacking. Meanwhile, management remains elusive. METHODS Medical records from all consecutive NSCLC patients who were treated with ICI from 2015 to 2018 were retrospectively analyzed. The HP incidence rate was calculated according to five definitions (tumor growth rate [TGR]ratio, ΔTGR, tumor growth kinetic [TGK], RECIST, and time to treatment failure [TTF]), and the agreement between such definitions was determined. The HP impact on overall survival (OS) was then assessed. The association between HP (defined using the TGRratio definition) and clinical and biological variables was also assessed. Clinical HP management and its impact on outcomes were described. RESULTS We identified 169 consecutive ICI-treated patients, with potential HP accounting for 11.3 %, 5.7 %, 17.0 %, 9.6 %, and 31.7 % patients, according to TGRratio, ΔTGR, TGK, RECIST, and TTF definitions. Agreement between the different HP definitions was highly heterogeneous (range 29 %-77 %) and globally poor. HP was associated with shorter OS, compared to standard RECIST progressive disease, but this difference only reached statistical significance when using the TTF definition. TGRratio-based HP was significantly associated with hepatic metastases. In TGRratio-based HP patients, neither resuming chemotherapy nor corticosteroids use was associated with statistically significant impact on overall survival. CONCLUSION We found fairly heterogeneous HP rates using different definitions. TTF was the only definition leading to significantly worsened OS. Further studies are needed to provide consensus recommendations for the assessment, definition, and management of HP, whose existence is likely real.
Collapse
|
147
|
Hyper-Progressive Disease: The Potential Role and Consequences of T-Regulatory Cells Foiling Anti-PD-1 Cancer Immunotherapy. Cancers (Basel) 2020. [PMID: 33375291 DOI: 10.3390/cancers13010048.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Antibody-mediated disruption of the programmed cell death protein 1 (PD-1) pathway has brought much success to the fight against cancer. Nevertheless, a significant proportion of patients respond poorly to anti-PD-1 treatment. Cases of accelerated and more aggressive forms of cancer following therapy have also been reported. Termed hyper-progressive disease (HPD), this phenomenon often results in fatality, thus requires urgent attention. Among possible causes of HPD, regulatory T-cells (Tregs) are of suspect due to their high expression of PD-1, which modulates Treg activity. Tregs are a subset of CD4+ T-cells that play a non-redundant role in the prevention of autoimmunity and is functionally dependent on the X chromosome-linked transcription factor FoxP3. In cancer, CD4+FoxP3+ Tregs migrate to tumors to suppress anti-tumor immune responses, allowing cancer cells to persist. Hence, Treg accumulation in tumors is associated with poor prognosis. In mice, the anti-tumor efficacy of anti-PD-1 can be enhanced by depleting Tregs. This suggests Tregs pose resistance to anti-PD-1 therapy. In this article, we review the relevant Treg functions that suppress tumor immunity and the potential effects anti-PD-1 could have on Tregs which are counter-productive to the treatment of cancer, occasionally causing HPD.
Collapse
|
148
|
Tay C, Qian Y, Sakaguchi S. Hyper-Progressive Disease: The Potential Role and Consequences of T-Regulatory Cells Foiling Anti-PD-1 Cancer Immunotherapy. Cancers (Basel) 2020; 13:E48. [PMID: 33375291 PMCID: PMC7796137 DOI: 10.3390/cancers13010048] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/18/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022] Open
Abstract
Antibody-mediated disruption of the programmed cell death protein 1 (PD-1) pathway has brought much success to the fight against cancer. Nevertheless, a significant proportion of patients respond poorly to anti-PD-1 treatment. Cases of accelerated and more aggressive forms of cancer following therapy have also been reported. Termed hyper-progressive disease (HPD), this phenomenon often results in fatality, thus requires urgent attention. Among possible causes of HPD, regulatory T-cells (Tregs) are of suspect due to their high expression of PD-1, which modulates Treg activity. Tregs are a subset of CD4+ T-cells that play a non-redundant role in the prevention of autoimmunity and is functionally dependent on the X chromosome-linked transcription factor FoxP3. In cancer, CD4+FoxP3+ Tregs migrate to tumors to suppress anti-tumor immune responses, allowing cancer cells to persist. Hence, Treg accumulation in tumors is associated with poor prognosis. In mice, the anti-tumor efficacy of anti-PD-1 can be enhanced by depleting Tregs. This suggests Tregs pose resistance to anti-PD-1 therapy. In this article, we review the relevant Treg functions that suppress tumor immunity and the potential effects anti-PD-1 could have on Tregs which are counter-productive to the treatment of cancer, occasionally causing HPD.
Collapse
Affiliation(s)
- Christopher Tay
- Immunology Frontier Research Center, Department of Experimental Immunology, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan; (C.T.); (Y.Q.)
| | - Yamin Qian
- Immunology Frontier Research Center, Department of Experimental Immunology, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan; (C.T.); (Y.Q.)
| | - Shimon Sakaguchi
- Immunology Frontier Research Center, Department of Experimental Immunology, Osaka University, Yamadaoka, Suita, Osaka 565-0871, Japan; (C.T.); (Y.Q.)
- Laboratory of Experimental Immunology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
149
|
Kim KH, Hur JY, Koh J, Cho J, Ku BM, Koh JY, Sun JM, Lee SH, Ahn JS, Park K, Ahn MJ, Shin EC. Immunological Characteristics of Hyperprogressive Disease in Patients with Non-small Cell Lung Cancer Treated with Anti-PD-1/PD-L1 Abs. Immune Netw 2020; 20:e48. [PMID: 33425433 PMCID: PMC7779871 DOI: 10.4110/in.2020.20.e48] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/24/2020] [Accepted: 12/08/2020] [Indexed: 12/13/2022] Open
Abstract
Hyperprogressive disease (HPD) is a distinct pattern of progression characterized by acceleration of tumor growth after treatment with anti-PD-1/PD-L1 Abs. However, the immunological characteristics have not been fully elucidated in patients with HPD. We prospectively recruited patients with metastatic non-small cell lung cancer treated with anti-PD-1/PD-L1 Abs between April 2015 and April 2018, and collected peripheral blood before treatment and 7-days post-treatment. HPD was defined as ≥2-fold increase in both tumor growth kinetics and tumor growth rate between pre-treatment and post-treatment. Peripheral blood mononuclear cells were analyzed by multi-color flow cytometry to phenotype the immune cells. Of 115 patients, 19 (16.5%) developed HPD, 52 experienced durable clinical benefit (DCB; partial response or stable disease ≥6 months), and 44 experienced non-hyperprogressive progression (NHPD). Patients with HPD had significantly lower progression-free survival (p<0.001) and overall survival (p<0.001). When peripheral blood immune cells were examined, the pre-treatment frequency of CD39+ cells among CD8+ T cells was significantly higher in patients with HPD compared to those with NHPD, although it showed borderline significance to predict HPD. Other parameters regarding regulatory T cells or myeloid derived suppressor cells did not significantly differ among patient groups. Our findings suggest high pre-treatment frequency of CD39+CD8+ T cells might be a characteristic of HPD. Further investigations in a larger cohort are needed to confirm our results and better delineate the immune landscape of HPD.
Collapse
Affiliation(s)
- Kyung Hwan Kim
- Department of Radiation Oncology, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul 03722, Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Joon Young Hur
- Division of Hematology and Oncology, Department of Internal Medicine, Hanyang University Guri Hospital, Guri 11923, Korea
| | - Jiae Koh
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06355, Korea
| | - Jinhyun Cho
- Division of Hematology-Oncology, Department of Internal Medicine, Inha University Hospital, Inha University School of Medicine, Incheon 22332, Korea
| | - Bo Mi Ku
- Research Institute for Future Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - June Young Koh
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Jong-Mu Sun
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Keunchil Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Myung-Ju Ahn
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06355, Korea.,Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| |
Collapse
|
150
|
Li J, Yan J, Cao R, Du G, Zhao G. Lung Adenocarcinoma Harboring EGFR Kinase Domain Duplication ( EGFR-KDD) Confers Sensitivity to Osimertinib and Nivolumab: A Case Report. Front Oncol 2020; 10:575739. [PMID: 33392076 PMCID: PMC7773813 DOI: 10.3389/fonc.2020.575739] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/12/2020] [Indexed: 11/13/2022] Open
Abstract
Background Kinase domain duplication of EGFR (EGFR-KDD) is a rare oncogenic driver alteration and serves as a potential therapeutic target. Its effect on EGFR-tyrosine kinase inhibitors (TKIs), especially the third-generation drug Osimertinib, and immune checkpoint inhibitors (ICIs) remains inconclusive. Case Presentation A 45-year old male with lung adenocarcinoma progressed with liver metastasis after receiving pemetrexed and cisplatin as adjuvant chemotherapy. Targeted next-generation sequencing (NGS) identified an EGFR-KDD in the resected left upper lung. Icotinib was used in the following treatment and the liver metastasis was found to shrink but the progression-free survival (PFS) only lasted for 4 months with the appearance of right hepatic metastasis. Meantime, the same EGFR-KDD was identified in the left hepatic re-biopsy. Afterward, the patient benefited from the third-line therapy of Osimertinib with a PFS as long as 21 months. Then he progressed with enlarged mediastinal lymph nodes, and targeted NGS consistently identified EGFR-KDD, as well as a new RELN p.G1774E mutation. Given the continually increasing tumor mutation burden (TMB, 3.4 mutation/Mb) and PD-L1 expression-based tumor proportion score (TPS, 1%), Nivolumab was used as the fourth-line salvage therapy, which lead to considerable efficacy, with decreased blood carcinoembryonic antigen (CEA), regressed mediastinal lymph nodes, and reduced liver metastases. Conclusions Our case provided direct evidence to support the role of Osimertinib in the treatment of EGFR-KDD, as well as added valuable insights into application of immune-based therapeutics in the specific subgroups bearing EGFR alteration(s).
Collapse
Affiliation(s)
- Jie Li
- Department of Thoracic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Junrong Yan
- Medical Department, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Ran Cao
- Translational Medicine Research Institute, Geneseeq Technology Inc., Toronto, ON, Canada
| | - Guanjun Du
- Medical Department, Nanjing Geneseeq Technology Inc., Nanjing, China
| | - Guofang Zhao
- Department of Thoracic Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| |
Collapse
|