1
|
Şen GA, Öztaş NŞ, Değerli E, Guliyev M, Can G, Turna H, Özgüroğlu M. Hyperprogressive disease in patients with advanced cancer treated with immune checkpoint inhibitors. Clin Transl Oncol 2024; 26:3264-3271. [PMID: 39225958 DOI: 10.1007/s12094-024-03696-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Hyperprogressive disease (HPD) is a new phenomenon developing in the era of immune checkpoint inhibitor (ICI) therapy. HPD is characterized by an unexpected and fast progression in tumor volume and poor survival. There is no standardized definition for HPD and clinicopathological variables associated with HPD are unclear. Herein, we assessed incidence, treatment outcomes and factors predictive of HPD in patients treated with ICIs. METHODS We retrospectively analyzed patients with advanced cancer treated with ICI at one academic center between 2014 and 2021. We used the Lo Russo's adopted criteria combined with clinical and radiologic parameters for the definition of HPD. All patients who underwent their first tumor evaluation according to RECIST1.1 were included. RESULTS Of 155 patients, 147 were eligible for analysis. The median age was 61 and 83% were male. The cancer types were; lung 67.3%, bladder 12.9%, gastric 9.5%, 5, colon 5.4% and renal cell carcinoma 4.8%. 59.9% of patients were treatment-naive and others had one or more lines of chemotherapy. 19 (12.9%) patients had HPD. In patients who had HPD, progression-free survival (PFS) was significantly shorter (1.5 vs 9.8 months, (HR 9.56; 95% CI (5.51-16.57), p < 0.001). The median overall survival (OS) was also shorter for HPD patients than non-HPD (3.0 vs 23.1 months, respectively, HR 12.03, 95% CI (6.64-21.81), p < 0.001). Gastric cancer, larger sum of target lesion diameters at baseline, liver metastases, higher LDH level and higher neutrophil-lymphocyte ratio (NLR) were significantly associated with HPD. CONCLUSION Our findings demonstrated that HPD was a rapid phenomenon with significantly poor survival rates. Several clinicopathological factors and tumor characteristics might indicate HPD.
Collapse
Affiliation(s)
- Gülin Alkan Şen
- Division of Medical Oncology, Department of Internal Medicine, Cerrahpaşa Faculty of Medicine, İstanbul University-Cerrahpaşa, Istanbul, Turkey.
| | - Nihan Şentürk Öztaş
- Division of Medical Oncology, Department of Internal Medicine, Cerrahpaşa Faculty of Medicine, İstanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Ezgi Değerli
- Division of Medical Oncology, Department of Internal Medicine, Cerrahpaşa Faculty of Medicine, İstanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Murad Guliyev
- Division of Medical Oncology, Department of Internal Medicine, Cerrahpaşa Faculty of Medicine, İstanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Günay Can
- Department of Public Health, Cerrahpaşa Faculty of Medicine, İstanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Hande Turna
- Division of Medical Oncology, Department of Internal Medicine, Cerrahpaşa Faculty of Medicine, İstanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Mustafa Özgüroğlu
- Division of Medical Oncology, Department of Internal Medicine, Cerrahpaşa Faculty of Medicine, İstanbul University-Cerrahpaşa, Istanbul, Turkey
| |
Collapse
|
2
|
Naulleau G, Monnet I, Rousseau-Bussac G, Vinas F, Mangiapan G, Jabot L, Boudjemaa A, Chouaïd C, Auliac JB, Assié JB. Double immune checkpoint inhibitor therapy for unresectable pleural mesothelioma rarely induces hyperprogressive disease: a case report. Transl Lung Cancer Res 2024; 13:2448-2452. [PMID: 39430317 PMCID: PMC11484718 DOI: 10.21037/tlcr-24-382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/26/2024] [Indexed: 10/22/2024]
Abstract
Background Use of immune checkpoint inhibitors (ICIs) is associated with new response types, such as hyperprogressive disease (HPD), whose definition is still being discussed. Some authors use dynamic indexes to define HPD. However, since the Checkmate-743 study, ICIs have been a first-line therapy for pleural mesothelioma (PM), thereby making use of dynamic indexes less appropriate. The aim of this study is to describe two cases of HPD and then discuss its definitions and implications. Case Description Herein, we report two cases of PM HPD on first-line ICI therapy. A 67-year-old man with right unresectable epithelioid PM, without BAP1 or CDKN2A losses, high neutrophil/lymphocyte ratio and rapid-onset pulmonary and mediastinal HPD after two ICI cycles, died of respiratory failure 1 month after starting treatment. A 40-year-old woman with left unresectable epithelioid PM had HPD at first assessment after 4 ICI infusions with jugular thrombosis, liver metastases and more dismal biological parameters. There are multiple different ways to describe HPD, some not applicable to PM. Suspected mechanisms include macrophage reprogramming to M2 cells. There are no known predictive factors of HPD, and future works should focus on identifying them. Conclusions HPD is a mode of progression for ICI-treated PM patients. Further investigation is needed to better define and anticipate HPD in these patients.
Collapse
Affiliation(s)
- Gaspard Naulleau
- Department of Pneumology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Isabelle Monnet
- Department of Pneumology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | | | - Florent Vinas
- Department of Pneumology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Gilles Mangiapan
- Department of Pneumology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Laurence Jabot
- Department of Pneumology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Amel Boudjemaa
- Department of Pneumology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Christos Chouaïd
- Department of Pneumology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
- Department of Pneumology, Université de Paris-Est Créteil, Créteil, France
| | - Jean-Bernard Auliac
- Department of Pneumology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Jean-Baptiste Assié
- Department of Pneumology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
- Department of Pneumology, Université de Paris-Est Créteil, Créteil, France
- Centre de Recherche des Cordeliers, Inserm, Sorbonne Université, Université Paris Cité, Functional Genomics of Solid Tumors Laboratory, Paris, France
| |
Collapse
|
3
|
Alzeeb G, Tortorelli C, Taleb J, De Luca F, Berge B, Bardet C, Limagne E, Brun M, Chalus L, Pinteur B, Bravetti P, Gongora C, Apetoh L, Ghiringhelli F. Efficacy of novel allogeneic cancer cells vaccine to treat colorectal cancer. Front Oncol 2024; 14:1427428. [PMID: 39114302 PMCID: PMC11303197 DOI: 10.3389/fonc.2024.1427428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Colorectal cancer (CRC) remains a significant global health burden, emphasizing the need for innovative treatment strategies. 95% of the CRC population are microsatellite stable (MSS), insensitive to classical immunotherapies such as anti-PD-1; on the other hand, responders can become resistant and relapse. Recently, the use of cancer vaccines enhanced the immune response against tumor cells. In this context, we developed a therapeutic vaccine based on Stimulated Tumor Cells (STC) platform technology. This vaccine is composed of selected tumor cell lines stressed and haptenated in vitro to generate a factory of immunogenic cancer-related antigens validated by a proteomic cross analysis with patient's biopsies. This technology allows a multi-specific education of the immune system to target tumor cells harboring resistant clones. Here, we report safety and antitumor efficacy of the murine version of the STC vaccine on CT26 BALB/c CRC syngeneic murine models. We showed that one cell line (1CL)-based STC vaccine suppressed tumor growth and extended survival. In addition, three cell lines (3CL)-based STC vaccine significantly improves these parameters by presenting additional tumor-related antigens inducing a multi-specific anti-tumor immune response. Furthermore, proteomic analyses validated that the 3CL-based STC vaccine represents a wider quality range of tumor-related proteins than the 1CL-based STC vaccine covering key categories of tumor antigens related to tumor plasticity and treatment resistance. We also evaluated the efficacy of STC vaccine in an MC38 anti-PD-1 resistant syngeneic murine model. Vaccination with the 3CL-based STC vaccine significantly improved survival and showed a confirmed complete response with an antitumor activity carried by the increase of CD8+ lymphocyte T cells and M1 macrophage infiltration. These results demonstrate the potential of this technology to produce human vaccines for the treatment of patients with CRC.
Collapse
Affiliation(s)
| | | | - Jaqueline Taleb
- Imthernat, Université Claude Bernard Lyon 1, Therapies and Immune REsponse in Cancers (TIRECs), Lyon, France
| | | | | | | | - Emeric Limagne
- Transfer Platform for Cancer Biology, Centre Georges François Leclerc, Dijon, France
| | | | | | | | | | - Céline Gongora
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université de Montpellier, Montpellier, France
| | - Lionel Apetoh
- Brown Center for Immunotherapy, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Francois Ghiringhelli
- Transfer Platform for Cancer Biology, Centre Georges François Leclerc, Dijon, France
| |
Collapse
|
4
|
Djunadi TA, Oh Y, Lee J, Yu J, Chung LIY, Lee Y, Kim L, Hong T, Lee S, Shah Z, Park JH, Yoon SM, Chae YK. Redefining Clinical Hyperprogression: The Incidence, Clinical Implications, and Risk Factors of Hyperprogression in Non-Small Cell Lung Cancer Treated with Immunotherapy. Clin Lung Cancer 2024; 25:365-375.e14. [PMID: 38644088 DOI: 10.1016/j.cllc.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 02/29/2024] [Accepted: 03/02/2024] [Indexed: 04/23/2024]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) may be associated with hyperprogressive disease (HPD). However, there is currently no standardized definition of HPD, with its risk factors and clinical implications remaining unclear. We investigated HPD in lung cancer patients undergoing immunotherapy, aiming to redefine HPD, identify risk factors, and assess its impact on survival. METHODS Clinical and radiologic data from 121 non-small cell lung cancer (NSCLC) patients with 136 immunotherapy cases were reviewed retrospectively. Three HPD definitions (Champiat et al., HPDc; Saâda-Bouzid et al., HPDs; and Ferrara et al., HPDf) were employed. Additionally, all new measurable lesions on the post-treatment CT scan were incorporated in measuring the sum of longest diameters (SLD) to define modified HPD (mHPD). RESULTS Among the 121 patients, 4 (3.3%) had HPDc, 11 (9.1%) had HPDs, and none had HPDf. Adding all new measurable lesions increased HPD incidence by 5%-10% across definitions. Multivariate analysis revealed significantly lower progression-free survival (PFS) and overall survival (OS) for patients with HPDc (HR 5.25, P = .001; HR 3.75, P = .015) and HPDs (HR 3.74, P < .001; HR 3.46, P < .001) compared to those without. Patients with mHPD showed similarly poor survival outcomes as HPD patients. Liver metastasis at diagnosis was associated with HPDs, and a high tumor burden correlated with HPDc. CONCLUSIONS The incidence and risk factors of HPD varied with different definitions, but mHPD identified more cases with poor outcomes. This comprehensive approach may enhance the identification of at-risk patients and lead to a better understanding of HPD in lung cancer during immunotherapy.
Collapse
Affiliation(s)
- Trie Arni Djunadi
- Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Internal Medicine, Richmond University Medical Centre, Staten Island, NY
| | - Youjin Oh
- Feinberg School of Medicine, Northwestern University, Chicago, IL; Department of Internal Medicine, John H. Stroger, Jr. Hospital of Cook County, Chicago, IL
| | - Jeeyeon Lee
- Feinberg School of Medicine, Northwestern University, Chicago, IL; School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jisang Yu
- Feinberg School of Medicine, Northwestern University, Chicago, IL
| | | | - Yeunho Lee
- Department of Pediatrics, University of Hawai'i, Honolulu, HI
| | - Leeseul Kim
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL
| | | | | | - Zunairah Shah
- Department of Hematology Oncology, Roswell Park Comprehensive Care Center, Buffalo, NY
| | - Joo Hee Park
- Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Sung Mi Yoon
- Department of Internal Medicine, Jacobi Medical Center/North Central Bronx Hospital Albert Einstein College of Medicine, Bronx, NY
| | - Young Kwang Chae
- Feinberg School of Medicine, Northwestern University, Chicago, IL.
| |
Collapse
|
5
|
Kim MJ, Hong SPD, Park Y, Chae YK. Incidence of immunotherapy-related hyperprogressive disease (HPD) across HPD definitions and cancer types in observational studies: A systematic review and meta-analysis. Cancer Med 2024; 13:e6970. [PMID: 38400685 PMCID: PMC10891462 DOI: 10.1002/cam4.6970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 01/02/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND While evidence of hyperprogressive disease (HPD) continues to grow, the lack of a consensual definition obscures a proper characterization of HPD incidence. We examined how HPD incidence varies by the tumor type or the type of definition used. METHODS We searched PubMed, Embase, the Cochrane Library of Systematic Reviews, and Web of Science from database inception to June 21, 2022. Observational studies reporting HPD incidence, in patients diagnosed with solid malignant tumors and treated with immune checkpoint inhibitors (ICI), were included. Random-effects meta-analyses were performed, and all statistical tests were 2-sided. RESULTS HPD incidence was 12.4% (95% CI 10.2%-15.0%) with evidence of heterogeneity (Q = 119.32, p < 0.001). Meta-regression showed that the risk of developing HPD was higher in patients with advanced gastric cancer (adjusted odds ratio [OR], 10.83; 95% CI, 2.14-54.65; p < 0.001), hepatocellular carcinoma (adjusted OR, 7.99; 95% CI, 1.68-38.13; p = 0.006), non-small cell lung cancer (adjusted OR, 7.14; 95% CI, 1.58-32.29; p = 0.005), and mixed or other types (adjusted OR, 5.09; 95% CI, 1.12-23.14, p = 0.018) than in patients with renal cell carcinoma. Across definitions, HPD defined as a tumor growth kinetics ratio ≥ 2 (adjusted OR, 1.82; 95% CI, 1.08-3.07; p = 0.025) based on the Response Evaluation Criteria in Solid Tumors (RECIST) reported higher incidence than when HPD was defined as RECIST-defined progressive disease and a change in the tumor growth rate (TGR) exceeding 50% (∆TGR > 50). CONCLUSIONS The incidence of immunotherapy-related HPD may vary across tumor types and definitions used, supporting the argument for a uniform and improved method of HPD evaluation for informed clinical decision-making.
Collapse
Affiliation(s)
- Min Jeong Kim
- Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Seung Pyo D. Hong
- Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Yeonggyeong Park
- Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Young Kwang Chae
- Department of MedicineNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
- Robert H. Lurie Comprehensive Cancer CenterNorthwestern UniversityChicagoIllinoisUSA
| |
Collapse
|
6
|
Lin Q, Wang X, Hu Y. The opportunities and challenges in immunotherapy: Insights from the regulation of PD-L1 in cancer cells. Cancer Lett 2023:216318. [PMID: 37454966 DOI: 10.1016/j.canlet.2023.216318] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
The immunosuppressive molecule programmed death-ligand 1 (PD-L1) is frequently upregulated in human cancers. Binding of PD-L1 to its receptor, programmed death-1 (PD-1), on activated T cells facilitates cancer cells to evade the host immune system. Antibody-based PD-1/PD-L1 inhibitors can inhibit PD-1/PD-L1 interaction allowing reactivate cytotoxic T cells to eradicate advanced cancer cells. However, the majority of cancer patients fail to respond to anti-PD-1/PD-L1 therapies and the molecular mechanisms for this remain poorly understood. Recent studies show that PD-L1 expression level on tumor cells affect the clinical efficacy of immune checkpoint therapies. Thus, furthering our understanding of the regulatory mechanisms of PD-L1 expression in cancer cells will be critical to improve clinical response rates and the efficacy of PD-1/PD-L1 immune therapies. Here we review recent studies, primarily focusing on the mechanisms that regulate PD-L1 expression at the transcriptional, post-transcriptional and protein level, with the purpose to drive the development of more targeted and effective anti-PD-1/PD-L1 cancer therapies.
Collapse
Affiliation(s)
- Qingyu Lin
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, China
| | - Xingwen Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, China
| | - Ying Hu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang Province, 150001, China; Key Laboratory of Science and Engineering for the Multi-modal Prevention and Control of Major Chronic Diseases, Ministry of Industry and Information Technology, China.
| |
Collapse
|
7
|
Briukhovetska D, Suarez-Gosalvez J, Voigt C, Markota A, Giannou AD, Schübel M, Jobst J, Zhang T, Dörr J, Märkl F, Majed L, Müller PJ, May P, Gottschlich A, Tokarew N, Lücke J, Oner A, Schwerdtfeger M, Andreu-Sanz D, Grünmeier R, Seifert M, Michaelides S, Hristov M, König LM, Cadilha BL, Mikhaylov O, Anders HJ, Rothenfusser S, Flavell RA, Cerezo-Wallis D, Tejedo C, Soengas MS, Bald T, Huber S, Endres S, Kobold S. T cell-derived interleukin-22 drives the expression of CD155 by cancer cells to suppress NK cell function and promote metastasis. Immunity 2023; 56:143-161.e11. [PMID: 36630913 PMCID: PMC9839367 DOI: 10.1016/j.immuni.2022.12.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 09/28/2022] [Accepted: 12/13/2022] [Indexed: 01/12/2023]
Abstract
Although T cells can exert potent anti-tumor immunity, a subset of T helper (Th) cells producing interleukin-22 (IL-22) in breast and lung tumors is linked to dismal patient outcome. Here, we examined the mechanisms whereby these T cells contribute to disease. In murine models of lung and breast cancer, constitutional and T cell-specific deletion of Il22 reduced metastases without affecting primary tumor growth. Deletion of the IL-22 receptor on cancer cells decreases metastasis to a degree similar to that seen in IL-22-deficient mice. IL-22 induced high expression of CD155, which bound to the activating receptor CD226 on NK cells. Excessive activation led to decreased amounts of CD226 and functionally impaired NK cells, which elevated the metastatic burden. IL-22 signaling was also associated with CD155 expression in human datasets and with poor patient outcomes. Taken together, our findings reveal an immunosuppressive circuit activated by T cell-derived IL-22 that promotes lung metastasis.
Collapse
Affiliation(s)
- Daria Briukhovetska
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - Javier Suarez-Gosalvez
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - Cornelia Voigt
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - Anamarija Markota
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - Anastasios D. Giannou
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, and Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany,Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Maryam Schübel
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - Jakob Jobst
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - Tao Zhang
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, and Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Janina Dörr
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - Florian Märkl
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - Lina Majed
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - Philipp Jie Müller
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - Peter May
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - Adrian Gottschlich
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - Nicholas Tokarew
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - Jöran Lücke
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, and Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany,Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Arman Oner
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - Melanie Schwerdtfeger
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - David Andreu-Sanz
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - Ruth Grünmeier
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - Matthias Seifert
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - Stefanos Michaelides
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - Michael Hristov
- Institute for Cardiovascular Prevention (IPEK), University Hospital, Klinikum der Universität München, Munich, Germany
| | - Lars M. König
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | - Bruno Loureiro Cadilha
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany
| | | | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, Klinikum der Universität München, 80337 Munich, Germany
| | - Simon Rothenfusser
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany,Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), 85764 Neuherberg, Germany
| | - Richard A. Flavell
- Department of Immunobiology, School of Medicine, Yale University, New Haven, CT 06520, USA,Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Daniela Cerezo-Wallis
- Melanoma Laboratory, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Cristina Tejedo
- Melanoma Laboratory, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - María S. Soengas
- Melanoma Laboratory, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
| | - Tobias Bald
- Institute of Experimental Oncology, Medical Faculty, University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Samuel Huber
- Section of Molecular Immunology and Gastroenterology, I. Department of Medicine, and Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stefan Endres
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany,Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), 85764 Neuherberg, Germany,Center for Translational Cancer Research (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Klinikum der Universität München, 80337 Munich, Germany; Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), 85764 Neuherberg, Germany; Center for Translational Cancer Research (DKTK), Partner Site Munich, 80336 Munich, Germany.
| |
Collapse
|
8
|
Britt AS, Huang C, Huang CH. Hyperprogressive disease in non-small cell lung cancer treated with immune checkpoint inhibitor therapy, fact or myth? Front Oncol 2022; 12:996554. [DOI: 10.3389/fonc.2022.996554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 10/17/2022] [Indexed: 11/30/2022] Open
Abstract
The therapeutic landscape for patients with non-small cell lung cancer (NSCLC) has dramatically evolved with the development and adoption of immune checkpoint inhibitors (ICI) as front-line therapy. These novel antibodies target the interactions in immunoregulatory pathways, between programmed death-1 (PD-1) and programmed death ligand-1 (PD-L1), or cytotoxic T-lymphocyte antigen 4 (CTLA-4) and B7, resulting in the activation of T cells and cytotoxic response to induce an immunologic response. ICIs have demonstrated significant survival benefits and sustained responses in the treatment of NSCLC leading to the long-term survival of up to 5 year. One unusual response to ICI is a phenomenon termed Hyperprogressive Disease (HYD), which occurs in a subset of patients for whom ICI therapy can induce rapid disease growth, which ultimately leads to poorer outcomes with an incidence rate ranging from 5 to 37% in NSCLC patients. Prior reviews demonstrated that HYD can be defined by rapid tumor progression, deterioration of patient’s symptoms or new onset of disease. The mechanism of HYD could be related to genomic and tumor microenvironment changes and altered immune response. It will be important to establish a common definition of HYD for future research and clinical care.
Collapse
|
9
|
Wang MX, Gao SY, Yang F, Fan RJ, Yang QN, Zhang TL, Qian NS, Dai GH. Hyperprogression under treatment with immune-checkpoint inhibitors in patients with gastrointestinal cancer: A natural process of advanced tumor progression? World J Clin Oncol 2022; 13:729-737. [PMID: 36212599 PMCID: PMC9537503 DOI: 10.5306/wjco.v13.i9.729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/26/2022] [Accepted: 09/12/2022] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy has shown great promise in treating various types of malignant tumors. However, some patients with gastrointestinal cancer have been known to experience rapid disease progression after treatment, a situation referred to as hyperprogressive disease (HPD). This minireview focuses on the definitions and potential mechanisms of HPD, natural disease progression in gastrointestinal malignancies, and tumor immunological microenvironment.
Collapse
Affiliation(s)
- Mo-Xuan Wang
- Department of Oncology, Chinese PLA Medical School, Beijing 100853, China
| | - Shu-Yue Gao
- Department of Oncology, Chinese PLA Medical School, Beijing 100853, China
| | - Fan Yang
- Department of Oncology, Chinese PLA Medical School, Beijing 100853, China
| | - Run-Jia Fan
- Department of Oncology, Chinese PLA Medical School, Beijing 100853, China
| | - Qin-Na Yang
- Department of Oncology, Chinese PLA Medical School, Beijing 100853, China
| | - Tian-Lan Zhang
- Department of Oncology, Chinese PLA Medical School, Beijing 100853, China
| | - Nian-Song Qian
- Department of Oncology, Senior Department of Respiratory and Critical Care Medicine, The Eighth Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Guang-Hai Dai
- Department of Oncology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
10
|
Zhao Z, Bian J, Zhang J, Zhang T, Lu X. Hyperprogressive disease in patients suffering from solid malignancies treated by immune checkpoint inhibitors: A systematic review and meta-analysis. Front Oncol 2022; 12:843707. [PMID: 35992878 PMCID: PMC9381837 DOI: 10.3389/fonc.2022.843707] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 06/06/2022] [Indexed: 11/18/2022] Open
Abstract
Introduction Hyperprogressive disease (HPD) is a paradoxically rapid disease progression during or shortly after antitumor treatment, especially immune checkpoint inhibitors (ICIs). Various diagnosis criteria of HPD cause heterogeneous incidence rates in different clinical research, and there is no consensus on potential risk factors associated with HPD occurrence. Hence, we aimed to summarize incidence of HPD in ICI treatment for solid tumors. Clinicopathological factors associated with HPD are also analyzed. Methods Clinical studies about HPD during/after ICI treatment of solid malignancies are included. Pubmed, Embase, and Cochrane library were searched for eligible studies published before October 7. The Newcastle–Ottawa scale was used to assess the quality of the included studies. Random effect and fixed effect models were, respectively, used for pooling incidence of HPD and analysis of risk factors for HPD. Heterogeneity, subgroup analysis, and publication bias were also analyzed. All meta-analysis was performed via R software (y -40v4.0.2). Results Forty-one studies with 6009 patients were included. The pooled incidence of HPD was 13.2% (95% CI, 11.2%–15.4%). Head and neck cancer (HNC) had the highest incidence of HPD (18.06%), and melanoma had the lowest (9.9%). Tumor types (P = .0248) and gender ratio (P = .0116) are sources of heterogeneity of pooled incidence of HPD. For five clinicopathological factors associated with HPD, only programmed cell death protein 1 ligand 1 (PD-L1) positivity was a preventive factor (odds ratio = 0.61, P <.05). High lactate dehydrogenase (LDH) level (OR = 1.51, P = .01), metastatic sites >2 (OR = 2.38, P <.0001), Eastern Cooperative Oncology Group Performance Score ≥2 (OR = 1.47, P = .02), and liver metastasis (OR = 3.06, P <.0001) indicate higher risk of HPD. Conclusions The pooled incidence of HPD was less than 15%, and HNC had the highest incidence of HPD. LDH and PD-L1 are remarkable biomarkers for prediction of HPD in future medical practice.
Collapse
Affiliation(s)
| | | | | | | | - Xin Lu
- *Correspondence: Xin Lu, ;
| |
Collapse
|
11
|
Karasarides M, Cogdill AP, Robbins PB, Bowden M, Burton EM, Butterfield LH, Cesano A, Hammer C, Haymaker CL, Horak CE, McGee HM, Monette A, Rudqvist NP, Spencer CN, Sweis RF, Vincent BG, Wennerberg E, Yuan J, Zappasodi R, Lucey VMH, Wells DK, LaVallee T. Hallmarks of Resistance to Immune-Checkpoint Inhibitors. Cancer Immunol Res 2022; 10:372-383. [PMID: 35362046 PMCID: PMC9381103 DOI: 10.1158/2326-6066.cir-20-0586] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/15/2021] [Accepted: 01/24/2022] [Indexed: 01/29/2023]
Abstract
Immune-checkpoint inhibitors (ICI), although revolutionary in improving long-term survival outcomes, are mostly effective in patients with immune-responsive tumors. Most patients with cancer either do not respond to ICIs at all or experience disease progression after an initial period of response. Treatment resistance to ICIs remains a major challenge and defines the biggest unmet medical need in oncology worldwide. In a collaborative workshop, thought leaders from academic, biopharma, and nonprofit sectors convened to outline a resistance framework to support and guide future immune-resistance research. Here, we explore the initial part of our effort by collating seminal discoveries through the lens of known biological processes. We highlight eight biological processes and refer to them as immune resistance nodes. We examine the seminal discoveries that define each immune resistance node and pose critical questions, which, if answered, would greatly expand our notion of immune resistance. Ultimately, the expansion and application of this work calls for the integration of multiomic high-dimensional analyses from patient-level data to produce a map of resistance phenotypes that can be utilized to guide effective drug development and improved patient outcomes.
Collapse
Affiliation(s)
- Maria Karasarides
- Worldwide Medical Oncology, Bristol Myers Squibb, Princeton, New Jersey.,Corresponding Authors: Maria Karasarides, Worldwide Medical Oncology, Bristol-Myers Squibb, Boston, MA 021273401. E-mail: ; and Theresa LaVallee, 1 Letterman Drive, Suite D3500, San Francisco, CA 94129. Phone: 628-899-7593; E-mail:
| | - Alexandria P. Cogdill
- Immunai, New York, New York.,Department of Immunology, The University of Texas MD Anderson, Houston, Texas
| | | | - Michaela Bowden
- Translational Medicine, Bristol Myers Squibb, Cambridge, Massachusetts
| | - Elizabeth M. Burton
- Department of Surgical Oncology, The University of Texas MD Anderson, Houston, Texas
| | - Lisa H. Butterfield
- Parker Institute for Cancer Immunotherapy, San Francisco, California.,Department of Microbiology and Immunology, University of California San Francisco, San Francisco, California
| | | | - Christian Hammer
- Department of Cancer Immunology, Genentech, South San Francisco, California.,Department of Human Genetics, Genentech, South San Francisco, California
| | - Cara L. Haymaker
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Christine E. Horak
- Global Drug Development, Bristol Myers Squibb, Lawrenceville, New Jersey
| | - Heather M. McGee
- Department of Radiation Oncology, City of Hope National Medical Center and Department of Immuno-Oncology, Beckmann Research Institute, City of Hope, Duarte, California
| | - Anne Monette
- Lady Davis Institute for Medical Research, Montréal, Québec, Canada
| | | | - Christine N. Spencer
- Department of Informatics, Parker Institute for Cancer Immunotherapy, San Francisco, California.,University of California San Francisco, San Francisco, California
| | - Randy F. Sweis
- Department of Medicine, Section of Hematology/Oncology, University of Chicago, Chicago, Illinois.,Committee on Immunology, University of Chicago, Chicago, Illinois.,Comprehensive Cancer Center, University of Chicago, Chicago, Illinois
| | - Benjamin G. Vincent
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina
| | | | - Jianda Yuan
- Translational Oncology, Early Oncology Development Department, Merck Research Laboratories, Rahway, New Jersey
| | - Roberta Zappasodi
- Weill Cornell Medicine, New York, New York.,Ludwig Collaborative and Swim Across America Laboratory, Memorial Sloan Kettering Cancer Center, New York, New York.,Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Daniel K. Wells
- Immunai, New York, New York.,Parker Institute for Cancer Immunotherapy, San Francisco, California
| | - Theresa LaVallee
- Parker Institute for Cancer Immunotherapy, San Francisco, California.,Corresponding Authors: Maria Karasarides, Worldwide Medical Oncology, Bristol-Myers Squibb, Boston, MA 021273401. E-mail: ; and Theresa LaVallee, 1 Letterman Drive, Suite D3500, San Francisco, CA 94129. Phone: 628-899-7593; E-mail:
| |
Collapse
|
12
|
Cevik M, Namal E, Iner-Koksal U, Dinc-Sener N, Karaalp A, Ciftci C, Susleyici B. Association of PD-1 and PDL-1 gene polymorphisms with colorectal cancer risk and prognosis. Mol Biol Rep 2022; 49:1827-1836. [PMID: 35076848 DOI: 10.1007/s11033-021-06992-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/19/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Programmed Cell Death-1 (PD-1) together with Programmed Death Ligand 1 (PDL-1) have crucial roles in anti-tumor immune response, cancer susceptibility and prognosis. Since PD-1 and PDL-1 have been considered as important genetic risk factors in cancer development and their functions can be affected by polymorphic sites, we investigated the effects of PD-1 rs2227981, rs2227982, rs36084323 and PDL-1 rs2282055, rs822336 gene polymorphisms on colorectal cancer (CRC) risk and prognosis in Turkish subjects. METHODS AND RESULTS Our study group consisted of 5-FU or Capacitabine prescribed CRC diagnosed patients and healthy controls. Genotype analyses of PD1 and PDL-1 polymorphisms were performed with Agena MassARRAY platform. rs36084323 CT genotype frequency was found to be higher in controls compared to cases (p < 0.001). rs36084323 CT genotype was highly associated with reduced CRC risk compared to CC genotype (OR 0.068, 95% CI 0.022-0.211, p < 0.001). In adjusted analysis, rs2282055 GG genotype was found to be associated with reduced CRC risk (OR 0.271, 95% CI 0.078-0.940, p = 0.040). rs2282055 TT genotype was found to be related to longer progression-free (Bonferroni corrected Log rank p = 0.013) and overall survival (Bonferroni corrected Log rank p = 0.009) to that of GG genotypes. Patients with rs822336 GC+CC genotypes showed longer overall survival times compared to GG (Log rank p = 0.044). CONCLUSIONS According to our results, PD-1 rs822336 G > C polymorphism might be useful in predicting CRC prognosis. PDL-1 rs2282055 T > G polymorphism might be useful in predicting both CRC risk and prognosis. Further studies should be conducted in larger and different populations to clear the roles of PD-1 and PDL-1 polymorphisms in CRC risk and prognosis.
Collapse
Affiliation(s)
- Mehtap Cevik
- Department of Molecular Biology, Marmara University Faculty of Arts and Science, Istanbul, Turkey
| | - Esat Namal
- Department of Medical Oncology, Demiroglu Bilim University Faculty of Medicine, Istanbul, Turkey
| | | | - Nur Dinc-Sener
- Department of Medical Oncology, Demiroglu Bilim University Faculty of Medicine, Istanbul, Turkey
| | - Atila Karaalp
- Department of Medical Pharmacology, Marmara University School of Medicine, Istanbul, Turkey
| | - Cavlan Ciftci
- Department of Cardiology, Demiroglu Bilim University Faculty of Medicine, Istanbul, Turkey
| | - Belgin Susleyici
- Department of Molecular Biology, Marmara University Faculty of Arts and Science, Istanbul, Turkey.
| |
Collapse
|
13
|
Brest P, Mograbi B, Hofman P, Milano G. COVID-19 vaccination and cancer immunotherapy: should they stick together? Br J Cancer 2022; 126:1-3. [PMID: 34799696 PMCID: PMC8603902 DOI: 10.1038/s41416-021-01618-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 09/29/2021] [Accepted: 10/27/2021] [Indexed: 02/08/2023] Open
Abstract
The combination of COVID-19 vaccination with immunotherapy by checkpoint inhibitors in cancer patients could intensify immunological stimulation with potential reciprocal benefits. Here, we examine more closely the possible adverse events that can arise in each treatment modality. Our conclusion is that caution should be exercised when combining both treatments.
Collapse
Affiliation(s)
- Patrick Brest
- grid.417812.90000 0004 0639 1794Université Côte d’Azur, CNRS, Inserm, IRCAN, Centre Antoine Lacassagne, FHU-OncoAge, 06189 Nice, France
| | - Baharia Mograbi
- grid.417812.90000 0004 0639 1794Université Côte d’Azur, CNRS, Inserm, IRCAN, Centre Antoine Lacassagne, FHU-OncoAge, 06189 Nice, France
| | - Paul Hofman
- grid.417812.90000 0004 0639 1794Université Côte d’Azur, CNRS, Inserm, IRCAN, Centre Antoine Lacassagne, FHU-OncoAge, 06189 Nice, France ,grid.460782.f0000 0004 4910 6551Université Côte d’Azur, Laboratory of Clinical and Experimental Pathology, CHU-Nice, FHU OncoAge, Hospital-Integrated Biobank (BB-0033-00025), Nice, France
| | - Gerard Milano
- Université Côte d'Azur, Centre Antoine Lacassagne, EA7497, Nice, France.
| |
Collapse
|
14
|
Park HJ, Qin L, Bakouny Z, Krajewski KM, Van Allen EM, Choueiri TK, Shinagare AB. OUP accepted manuscript. Oncologist 2022; 27:389-397. [PMID: 35348767 PMCID: PMC9074990 DOI: 10.1093/oncolo/oyac034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Accepted: 01/07/2022] [Indexed: 11/15/2022] Open
Abstract
Background Materials and Methods Results Conclusion
Collapse
Affiliation(s)
- Hyo Jung Park
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Lei Qin
- Department of Imaging, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ziad Bakouny
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Katherine M Krajewski
- Department of Imaging, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Toni K Choueiri
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Atul B Shinagare
- Department of Imaging, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Corresponding author: Atul B. Shinagare, Department of Radiology, Brigham and Womens Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA. Tel.: +1 6176322988; Fax: +1 6175828574;
| |
Collapse
|
15
|
Strazza M, Bukhari S, Tocheva AS, Mor A. PD-1-induced proliferating T cells exhibit a distinct transcriptional signature. Immunology 2021; 164:555-568. [PMID: 34164813 PMCID: PMC8517598 DOI: 10.1111/imm.13388] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 05/24/2021] [Accepted: 06/19/2021] [Indexed: 12/12/2022] Open
Abstract
Ligation of the inhibitory receptor PD-1 on T cells results in the inhibition of numerous cellular functions. Despite the overtly inhibitory outcome of PD-1 signalling, there are additionally a collection of functions that are activated. We have observed that CD4+ T cells stimulated through the T-cell receptor and PD-1 primarily do not proliferate; however, there is a population of cells that proliferates more than T-cell receptor stimulation alone. These highly proliferating cells could potentially be associated with PD-1-blockade unresponsiveness in patients. In this study, we have performed RNA sequencing and found that following PD-1 ligation proliferating and non-proliferating T cells have distinct transcriptional signatures. Remarkably, the proliferating cells showed an enrichment of genes associated with an activated state despite PD-1 signalling. Additionally, circulating follicular helper T cells were significantly more prevalent in the non-proliferating population, demonstrated by enrichment of the associated genes CXCR5, CCR7, TCF7, BCL6 and PRDM1 and validated at the protein level. Translationally, we also show that there are more follicular helper T cells in patients that respond favourably to PD-1 blockade. Overall, the presence of transcriptionally and functionally distinct T cell populations responsive to PD-1 ligation may provide insights into the clinical differences observed following therapeutic PD-1 blockade.
Collapse
Affiliation(s)
- Marianne Strazza
- Columbia Center for Translational ImmunologyColumbia University Medical CenterNew YorkNYUSA
| | - Shoiab Bukhari
- Columbia Center for Translational ImmunologyColumbia University Medical CenterNew YorkNYUSA
| | - Anna S. Tocheva
- Columbia Center for Translational ImmunologyColumbia University Medical CenterNew YorkNYUSA
| | - Adam Mor
- Columbia Center for Translational ImmunologyColumbia University Medical CenterNew YorkNYUSA
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
PD-L1 regulation revisited: impact on immunotherapeutic strategies. Trends Mol Med 2021; 27:868-881. [PMID: 34187739 DOI: 10.1016/j.molmed.2021.06.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/29/2021] [Accepted: 06/07/2021] [Indexed: 02/06/2023]
Abstract
A particularly promising cancer treatment is the use of monoclonal antibodies (mAbs) against immune checkpoints (i.e., immune checkpoint inhibitors; ICIs). However, many patients experience relapse and severe adverse events. To overcome these negative issues and improve efficiency, current approaches rely on combinatorial treatments, including some modulating the expression of programmed cell death receptor 1 (PD-1)/programmed death ligand 1 (PD-L1) immune checkpoints directly. In this review, we examine the recently discovered pathways involved in PD-L1 expression and highlight the relevant druggable strategies that are being developed to both improve the response rate and avoid the onset of resistance. Altogether, these new strategies will pave the way for effective treatment combinations in future oncology clinical trials.
Collapse
|
19
|
Minari R, Bonatti F, Mazzaschi G, Dodi A, Facchinetti F, Gelsomino F, Cinquegrani G, Squadrilli A, Bordi P, Buti S, Bersanelli M, Leonetti A, Cosenza A, Ferri L, Rapacchi E, Quaini F, Ardizzoni A, Tiseo M. PD-L1 SNPs as biomarkers to define benefit in patients with advanced NSCLC treated with immune checkpoint inhibitors. TUMORI JOURNAL 2021; 108:47-55. [PMID: 34002648 DOI: 10.1177/03008916211014954] [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] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To investigate the role of CTLA-4, PD-1 (programmed death-1), and PD-L1 (programmed death-ligand 1) single nucleotide polymorphisms (SNPs) in predicting clinical outcome of patients with advanced non-small cell lung cancer (NSCLC) treated with immune checkpoint inhibitors (ICIs). METHODS A total of 166 consecutive patients were included. We correlated SNPs with clinical benefit, progression-free survival, time to treatment failure, and overall survival and evaluated the incidence of SNPs in nonresponder and long clinical benefit groups. RESULTS Considering the entire cohort, no correlation was found between SNPs and clinical outcome; however, PD-L1 rs4143815 SNP and the long clinical benefit group showed a statistically significant association (p = 0.02). The nonresponder cohort displayed distinctive PD-L1 haplotype (p = 0.05). CONCLUSION PD-L1 SNPs seem to be marginally involved in predicting clinical outcome of NSCLC treated with ICI, but further investigations are required.
Collapse
Affiliation(s)
- Roberta Minari
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | | | - Giulia Mazzaschi
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy.,Department of Medicine & Surgery, University of Parma
| | | | - Francesco Facchinetti
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy.,Université Paris-Saclay, Institut Gustave Roussy, Inserm, Biomarqueurs Prédictifs et Nouvelles Stratégies Thérapeutiques en Oncologie, Villejuif, France
| | | | | | - Anna Squadrilli
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Paola Bordi
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Sebastiano Buti
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | | | | | - Agnese Cosenza
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Leonarda Ferri
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Elena Rapacchi
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | | | - Andrea Ardizzoni
- Medical Oncology, AOU Policlinico S. Orsola-Malpighi, Bologna, Italy
| | - Marcello Tiseo
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy.,Department of Medicine & Surgery, University of Parma
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
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
|
23
|
Heterogeneity of Response and Immune System Activity during Treatment with Nivolumab in Hepatocellular Carcinoma: Results from a Single-Institution Retrospective Analysis. Cancers (Basel) 2021; 13:cancers13020213. [PMID: 33430142 PMCID: PMC7827490 DOI: 10.3390/cancers13020213] [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: 12/07/2020] [Revised: 12/27/2020] [Accepted: 01/05/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Immunotherapy is an emerging treatment in hepatocellular carcinoma, both alone and in combination. The advent of this new approach raises challenges for the interpretation of response assessment due to the peculiar patterns of mixed responses, pseudoprogression and hyperprogression. Furthermore, there are no criteria to drive selection of treatment strategy. We analyzed data from the first 10 patients treated with nivolumab in our institution and we identified different patterns of response according to the lesion’s site. Furthermore, we analyzed blood samples from the first four patients, and found differences, between a patient with shorter survival and the other three, that may provide insight into mechanisms underlying the different activities of nivolumab. Although we analyzed data from a small number of patients, our results can help to understand mechanisms of immunotherapy activity in order to define the most appropriate treatment strategy for each patient. Abstract Treatment of hepatocellular carcinoma (HCC) is rapidly evolving, with many new therapeutic options; in particular, immunotherapy (IT) is acquiring a major role, even in combination regimens. Despite these promising results, an important limitation is the lack of prognostic and predictive factors that prevent provision of a tool for patient stratification in order to select the most appropriate strategy. Furthermore, response assessment can be challenging with IT due to peculiar patterns such as mixed responses or pseudoprogression. We analyzed biological and clinical features from the first 10 HCC patients treated with nivolumab in our institution. Analysis of patterns of response in CT assessment revealed complete response in pulmonary lesions, along with heterogeneous behavior in the liver and other organ lesions. Peripheral blood mononuclear cells (PBMC) analysis in the first four patients showed unique alterations in a patient with poor prognosis, both at baseline (lower percentage of effector T cells, higher percentage of natural killer T [NK/T] cells) and during treatment with nivolumab (decrease in nonclassical monocytes, increase in monocytic myeloid-derived suppressor cells [MO-MDSC]), suggesting a possible prognostic role for these features. Although obtained in a small cohort of patients, our results open a new perspective for understanding mechanisms underlying IT outcomes in HCC patients.
Collapse
|
24
|
Yoshida H, Nomizo T, Ozasa H, Tsuji T, Funazo T, Yasuda Y, Ajimizu H, Yamazoe M, Kuninaga K, Ogimoto T, Hosoya K, Itotani R, Sakamori Y, Kim YH, Hirai T. PD-L1 polymorphisms predict survival outcomes in advanced non-small-cell lung cancer patients treated with PD-1 blockade. Eur J Cancer 2020; 144:317-325. [PMID: 33385948 DOI: 10.1016/j.ejca.2020.11.035] [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: 08/21/2020] [Revised: 11/13/2020] [Accepted: 11/24/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND We previously reported that PD-L1 polymorphisms are associated with the efficacy and immune-related adverse events of PD-1 blockade with nivolumab. However, the association between PD-L1 polymorphisms and survival outcomes under PD-1/PD-L1 blockade is still uncertain. Here, we aimed to investigate whether PD-L1 polymorphisms are associated with survival outcomes in advanced non-small-cell lung cancer (NSCLC) patients treated with nivolumab. METHODS PD-1/PD-L1 polymorphisms and survival outcomes were retrospectively analysed in two independent cohorts (133 patients treated with nivolumab and 96 patients with no treatment history of an immune checkpoint inhibitor (ICI) (the non-ICI cohort)) with advanced NSCLC. RESULTS Among the 7 studied single-nucleotide polymorphisms, PD-L1 rs822339 and rs1411262 were associated with overall survival (OS) in patients treated with nivolumab. Patients with the A/A genotype of rs822339 had a significantly longer OS than those with A/G or G/G genotypes (not reached versus 12.0 months; hazard ratio (HR), 0.35; 95% confidence interval (CI), 0.18-0.64; p = 0.0008). A similar survival benefit with the A/A genotype was observed regardless of driver mutation status. In multivariate analysis, performance status (PS) and PD-L1 rs822339 genotype were independent prognostic factors for OS. In the non-ICI cohort, the PD-L1 rs822339 genotype did not correlate with OS (HR, 0.77; 95% CI, 0.31-1.70; p = 0.55). The T/T genotype of rs1411262 also showed a significant prolongation of OS compared to that with the C/T or C/C genotypes in patients treated with nivolumab. CONCLUSIONS PD-L1 polymorphisms are associated with favourable OS in nivolumab-treated NSCLC patients and may be useful predictive biomarkers, regardless of driver mutation status.
Collapse
Affiliation(s)
- Hironori Yoshida
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Japan
| | - Takashi Nomizo
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Japan
| | - Hiroaki Ozasa
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Japan.
| | - Takahiro Tsuji
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Japan
| | - Tomoko Funazo
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Japan
| | - Yuto Yasuda
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Japan
| | - Hitomi Ajimizu
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Japan
| | - Masatoshi Yamazoe
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Japan
| | - Kiyomitsu Kuninaga
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Japan
| | - Tatsuya Ogimoto
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Japan
| | - Kazutaka Hosoya
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Japan
| | - Ryo Itotani
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Japan
| | - Yuichi Sakamori
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Japan
| | - Young Hak Kim
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Japan
| |
Collapse
|
25
|
Toki MI, Syrigos N, Syrigos K. Hyperprogressive disease: A distinct pattern of progression to immune checkpoint inhibitors. Int J Cancer 2020; 149:277-286. [PMID: 33300601 DOI: 10.1002/ijc.33429] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 12/31/2022]
Abstract
Hyperprogressive disease (HPD) is a novel pattern of progression attributed to immune checkpoint inhibitor (ICI) treatment and characterized by a dramatic tumor surge and poor survival. The concept of HPD is still controversial, while the definition varies widely across studies. Although HPD has been associated with multiple clinicopathological and molecular features, there is no biomarker to predict this detrimental effect of immunotherapy and the underlying mechanism remains unknown. The aim of this comprehensive review is to summarize current data on HPD and present the controversies and clinical care management challenges for oncologists treating patients with ICIs.
Collapse
Affiliation(s)
- Maria I Toki
- Department of Pathology, Yale University, School of Medicine, New Haven, Connecticut, USA.,National and Kapodistrian University, School of Medicine, Athens, Greece
| | - Nikos Syrigos
- National and Kapodistrian University, School of Medicine, Athens, Greece
| | - Kostas Syrigos
- National and Kapodistrian University, School of Medicine, Athens, Greece
| |
Collapse
|
26
|
Camelliti S, Le Noci V, Bianchi F, Moscheni C, Arnaboldi F, Gagliano N, Balsari A, Garassino MC, Tagliabue E, Sfondrini L, Sommariva M. Mechanisms of hyperprogressive disease after immune checkpoint inhibitor therapy: what we (don't) know. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:236. [PMID: 33168050 PMCID: PMC7650183 DOI: 10.1186/s13046-020-01721-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/25/2020] [Indexed: 02/07/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have made a breakthrough in the treatment of different types of tumors, leading to improvement in survival, even in patients with advanced cancers. Despite the good clinical results, a certain percentage of patients do not respond to this kind of immunotherapy. In addition, in a fraction of nonresponder patients, which can vary from 4 to 29% according to different studies, a paradoxical boost in tumor growth after ICI administration was observed: a completely unpredictable novel pattern of cancer progression defined as hyperprogressive disease. Since this clinical phenomenon has only been recently described, a universally accepted clinical definition is lacking, and major efforts have been made to uncover the biological bases underlying hyperprogressive disease. The lines of research pursued so far have focused their attention on the study of the immune tumor microenvironment or on the analysis of intrinsic genomic characteristics of cancer cells producing data that allowed us to formulate several hypotheses to explain this detrimental effect related to ICI therapy. The aim of this review is to summarize the most important works that, to date, provide important insights that are useful in understanding the mechanistic causes of hyperprogressive disease.
Collapse
Affiliation(s)
- Simone Camelliti
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, via Mangiagalli 31, 20133, Milan, Italy
| | - Valentino Le Noci
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, via Mangiagalli 31, 20133, Milan, Italy
| | - Francesca Bianchi
- Molecular Targets Unit, Department of Research, Fondazione IRCCS - Istituto Nazionale dei Tumori, via Amadeo 42, 20133, Milan, Italy
| | - Claudia Moscheni
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, via Mangiagalli 31, 20133, Milan, Italy
| | - Francesca Arnaboldi
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, via Mangiagalli 31, 20133, Milan, Italy
| | - Nicoletta Gagliano
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, via Mangiagalli 31, 20133, Milan, Italy
| | - Andrea Balsari
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, via Mangiagalli 31, 20133, Milan, Italy
| | - Marina Chiara Garassino
- Thoracic Oncology Unit, Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, via Venezian 1, 20133, Milan, Italy
| | - Elda Tagliabue
- Molecular Targets Unit, Department of Research, Fondazione IRCCS - Istituto Nazionale dei Tumori, via Amadeo 42, 20133, Milan, Italy
| | - Lucia Sfondrini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, via Mangiagalli 31, 20133, Milan, Italy
| | - Michele Sommariva
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, via Mangiagalli 31, 20133, Milan, Italy.
| |
Collapse
|
27
|
Choi YJ, Kim T, Kim EY, Lee SH, Kwon DS, Chang YS. Prediction model for hyperprogressive disease in non-small cell lung cancer treated with immune checkpoint inhibitors. Thorac Cancer 2020; 11:2793-2803. [PMID: 32779394 PMCID: PMC7529559 DOI: 10.1111/1759-7714.13594] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/08/2020] [Accepted: 07/09/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Hyperprogressive disease (HPD) is a paradoxical acceleration of tumor growth after immune checkpoint inhibitor (ICI) treatment. This study aimed to identify the risk factors and to present a predictive model for HPD in patients treated with ICIs. METHODS A total of 78 non-small cell lung cancer (NSCLC) cases, treated with at least two cycles of ICIs who underwent computed tomography (CT) for response assessment were recruited into the study from January 2016 to August 2019. HPD was defined by the following criteria: (i) time-to-treatment failure <2 months; (ii) a 50% increase in the sum of target lesion diameters; (iii) new development of at least two lesions in an already involved organ; (iv) appearance of a new organ lesion; and (v) a decrease in ECOG PS 2. RESULTS Of the 78 total patients, 15 (19.2%) had HPD. The risk factors of HPD were age; primary lesion size; and metastases in the contralateral lung, pleura, liver, and bone in multivariable logistic regression (odds ratio [OR]; 0.9038, 1.6619, 28.5913, 23.8264, 14.5711, and 20.1533, respectively, all P-values < 0.05). By using these risk factors, we developed a prediction model for HPD and the area under the receiver operating characteristic curve of the model was 0.9556 (95% confidence interval [CI]: 0.9133-0.9978). CONCLUSIONS HPD is relatively common and associated with a grave clinical outcome, requiring a careful monitoring in lung cancer patients treated with ICIs. Moreover, risk factors such as age, size of tumor and number of various metastatic lesions should be taken into consideration before ICI administration. KEY POINTS SIGNIFICANT FINDINGS OF THE STUDY: Age, primary lesion size, and number of metastases are risk factors of HPD. HPD is strongly associated with poor prognosis. HPD during ICI use needs comprehensive monitoring. WHAT THIS STUDY ADDS This is the first study to develop a prediction model. The area under the curve of the prediction model for HPD was 0.9556.
Collapse
Affiliation(s)
- Yong Jun Choi
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Taehee Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Eun Young Kim
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang Hoon Lee
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Do Sun Kwon
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Yoon Soo Chang
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
28
|
Park JH, Chun SH, Lee YG, Chang H, Lee KW, Kim HR, Shin SH, An HJ, Lee KE, Hwang IG, Ahn MJ, Kim SB, Keam B. Hyperprogressive disease and its clinical impact in patients with recurrent and/or metastatic head and neck squamous cell carcinoma treated with immune-checkpoint inhibitors: Korean cancer study group HN 18-12. J Cancer Res Clin Oncol 2020; 146:3359-3369. [PMID: 32671504 DOI: 10.1007/s00432-020-03316-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Although immune-checkpoint inhibitors (ICIs) have emerged as therapeutic options for recurrent and/or metastatic head and neck squamous cell carcinoma (R/M-HNSCC), concerns have been raised on exceptional acceleration of tumor growth during treatment with ICIs, a condition described as hyperprogressive disease (HPD). This study examined the incidence, potential predictors, and clinical impact of HPD in R/M-HNSCC. METHODS We retrospectively collected data of patients with R/M-HNSCC treated with ICIs between January 2013 and June 2018 from 11 medical centers in Korea. HPD was defined as tumor growth kinetics ratio (TGKr) > 2, which was calculated by comparing TGK on ICIs with that before treatment with ICIs. RESULTS Of 125 patients, 68 (54.4%) obtained progressive disease as their best responses (progressors). HPD was identified in 18 (26.5% of progressors, 14.4% of total) patients. Relatively younger age, primary tumor of oral cavity, and previous locoregional irradiation were significant predictors of HPD according to multivariable analysis (p = 0.040, 0.027, and 0.015, respectively). Compared to patients without HPD, patients with HPD had significantly shorter median progression-free survival (PFS) (1.2 vs. 3.4 months, p < 0.001) and overall survival (OS) (3.4 vs. 10.7 months, p = 0.047). However, interestingly, HPD did not significantly affect the therapeutic benefit of post-ICIs chemotherapy. CONCLUSIONS Younger patients with oral cavity cancer or prior treatment with locoregional radiotherapy could be regarded potential risk groups for HPD in patients with R/M-HNSCC treated with ICIs. Although HPD could consistently predict poorer survival outcomes, patients who experienced HPD with ICIs should not be excluded from the subsequent salvage chemotherapy treatments.
Collapse
Affiliation(s)
- Ji Hyun Park
- Department of Internal Medicine, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul, Republic of Korea
| | - Sang Hoon Chun
- Department of Internal Medicine, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Yun-Gyoo Lee
- Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyun Chang
- Department of Internal Medicine, International St. Mary's Hospital, Catholic Kwandong University College of Medicine, Incheon, Republic of Korea
| | - Keun-Wook Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Hye Ryun Kim
- Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seong Hoon Shin
- Department of Internal Medicine, Kosin University Gospel Hospital, Kosin University College of Medicine, Busan, Republic of Korea
| | - Ho Jung An
- Department of Internal Medicine, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Republic of Korea
| | - Kyoung Eun Lee
- Department of Hematology and Oncology, Ewha Woman's University Hospital, Seoul, Republic of Korea
| | - In Gyu Hwang
- Department of Internal Medicine, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Myung-Ju Ahn
- Department of Internal Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sung-Bae Kim
- Department of Internal Medicine, Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, 88 Olympic-ro 43-gil, Songpa-gu, Seoul, 05505, Republic of Korea.
| | - Bhumsuk Keam
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
| |
Collapse
|