1
|
Yao S, Chen S, Wang A, Liang Z, Liu X, Gao Y, Cai H. BAG2 Inhibits Cervical Cancer Progression by Modulating Type I Interferon Signaling through Stabilizing STING. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e70005. [PMID: 40364789 DOI: 10.1002/advs.202414637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 04/30/2025] [Indexed: 05/15/2025]
Abstract
Cervical cancer possesses high morbidity and mortality rates, and a comprehensive understanding of its molecular underpinnings is essential for advancing clinical management strategies. The innate immune sensor STING, which activates type I interferon signaling, plays a pivotal role in enhancing anti-tumor activity. Despite increased attention to STING's involvement in cervical cancer, the regulatory mechanisms governing its protein homeostasis remain poorly understood. In this study, it is found that the BAG2-STUB1 complex regulates ubiquitin proteasomal degradation of STING, which affects the development of cervical cancer. Mechanistically, BAG2 inhibits ubiquitination of STING and stabilizes it by interacting with STING. Specifically, BAG2 inhibits STUB1 from attaching the K48-linked ubiquitin chains at K338 and K370 of STING by forming a complex with STUB1. Functionally, enhanced BAG2 expression suppresses cervical cancer progression by activating the type I interferon pathway in a STING-dependent manner. Notably, clinical cervical cancer samples revealed a positive correlation between BAG2 and STING levels, with low BAG2 expression is strongly linked to advanced disease and poor prognosis in cervical cancer. Collectively, these findings elucidate the molecular mechanism by which the BAG2-STUB1 complex regulates STING homeostasis, underscoring BAG2's potential as a diagnostic biomarker and therapeutic target in cervical cancer.
Collapse
Affiliation(s)
- Shijie Yao
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071, China
- Hubei Cancer Clinical Study Center, Wuhan, 430071, China
| | - Siming Chen
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Anjin Wang
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071, China
- Hubei Cancer Clinical Study Center, Wuhan, 430071, China
| | - Ziyan Liang
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071, China
- Hubei Cancer Clinical Study Center, Wuhan, 430071, China
| | - Xuelian Liu
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071, China
- Hubei Cancer Clinical Study Center, Wuhan, 430071, China
| | - Yang Gao
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071, China
- Hubei Cancer Clinical Study Center, Wuhan, 430071, China
| | - Hongbing Cai
- Department of Gynecological Oncology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, Hubei, China
- Hubei Key Laboratory of Tumor Biological Behaviors, Wuhan, 430071, China
- Hubei Cancer Clinical Study Center, Wuhan, 430071, China
| |
Collapse
|
2
|
Muğlu H, Sünger E, Mıldanoğlu MM, Engin Delipoyraz E, Yücel MH, Özçelik H, Hamdard J, Açıkgöz Ö, Ölmez ÖF, Yıldız Ö, Bilici A. Clinicopathological Characteristics of Extrapulmonary Neuroendocrine Carcinomas: Treatment Responses and Survival Outcomes: Single-Center Experience. J Clin Med 2025; 14:2264. [PMID: 40217714 PMCID: PMC11989432 DOI: 10.3390/jcm14072264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 03/18/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: Extrapulmonary neuroendocrine carcinomas (EP-NECs) are rare, aggressive malignancies with no standardized treatment approach. Although platinum-based chemotherapy is considered the first-line therapy, overall survival (OS) and progression-free survival (PFS) remain limited. This study aims to evaluate the clinical and pathological characteristics of EP-NEC patients, their treatment responses, and survival outcomes. Methods: This retrospective observational study included 29 EP-NEC patients diagnosed and followed between 2015 and 2024. Clinical and demographic data, tumor localization, disease stage, administered treatments, and survival outcomes were analyzed. Kaplan-Meier survival analysis was used to assess OS and PFS, with subgroup comparisons performed via the log-rank test. Results: The most common primary tumor sites were the pancreas (21%), prostate (17%), and cervix (14%). At diagnosis, 55.2% of patients had metastatic disease. First-line platinum-based chemotherapy achieved an objective response rate of 82.1%, with a median PFS of 8.16 months and a median OS of 14.16 months. Surgical intervention significantly improved survival (p = 0.020), while a high Ki-67 proliferation index (>80%) was associated with worse PFS (p = 0.032). Other factors, including smoking status and liver-directed therapies, had no significant impact on survival. Conclusions: EP-NECs present with a poor prognosis despite platinum-based chemotherapy achieving high response rates. Surgical resection improves survival outcomes, whereas high Ki-67 expression is associated with a worse prognosis. These findings highlight the need for further research into novel therapeutic strategies for EP-NECs.
Collapse
Affiliation(s)
- Harun Muğlu
- Department of Medical Oncology, Faculty of Medicine, Medipol University, Istanbul 34214, Türkiye; (E.S.); (M.M.M.); (E.E.D.); (M.H.Y.); (H.Ö.); (J.H.); (Ö.A.); (Ö.F.Ö.); (Ö.Y.); (A.B.)
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Li S, Feng M, Wang F, Liu D, Li M, Dai J, Yang Y, Chai Y, Chen W. Mycobacterium tuberculosis infection may increase the degrees of malignancy in lung adenocarcinoma. Front Immunol 2025; 16:1537520. [PMID: 40061944 PMCID: PMC11885956 DOI: 10.3389/fimmu.2025.1537520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 02/04/2025] [Indexed: 05/13/2025] Open
Abstract
Background The early diagnosis and management of lung adenocarcinoma co-existing with tuberculosis (LAC-TB) presents significant challenges in clinical settings. This is compounded by a paucity of robust clinical evidence elucidating the interactions between these two conditions. Methods This study included 14 patients diagnosed with LAC-TB, with an equal distribution among those with pulmonary tuberculosis (TB) and those with peripheral lymph node TB. Controls included patients with simple TB and those with lung adenocarcinoma (LAC). Histopathologic examinations confirmed typical changes in each group. Immunohistochemistry analyzed immune markers, focusing on PD-L1, while genomic analysis identified differential mutant genes. Results Pathological evaluations showed that LAC-TB and LAC groups expressed TTF-1 and Napsin A in their adenocarcinoma specimens. Notably, a higher proportion of patients in the LAC-TB group had a Ki-67 proliferation index of ≥10%. Subsequent Molecular analyses revealed significant differences in RALGAPA1 gene expression, with the LAC-TB group also exhibiting a greater median count of missense mutations, single nucleotide polymorphisms, and overall mutations, suggesting a higher malignancy level than the LAC group. Additionally, the LAC-TB group showed an increased tumor mutational burden, indicating a potentially better response to immunotherapy. Immunohistochemical assessments indicated that Mycobacterium tuberculosis (MTB) infection correlated with reduced infiltration of T cells and CD4+ T cells, alongside an upregulation of PD-L1 expression in LAC. Notably, PD-L1 was strongly expressed in the TB granuloma and surrounding areas. Conclusion Our findings suggest that MTB infection may increase the malignancy of LAC, with the pronounced expression of PD-L1 in granuloma regions constituting a pivotal mechanism underlying this relationship.
Collapse
Affiliation(s)
- Shanshan Li
- Department of Pathology, The Eighth Medical Center, General Hospital of the Chinese People's Liberation Army (PLA), Beijing, China
| | - Mengru Feng
- Department of Pathology, The Eighth Medical Center, General Hospital of the Chinese People's Liberation Army (PLA), Beijing, China
| | - Fenghua Wang
- Department of Pathology, The Eighth Medical Center, General Hospital of the Chinese People's Liberation Army (PLA), Beijing, China
| | - Dongxu Liu
- Department of Pathology, The Eighth Medical Center, General Hospital of the Chinese People's Liberation Army (PLA), Beijing, China
| | - Mingyan Li
- Department of Pathology, The Eighth Medical Center, General Hospital of the Chinese People's Liberation Army (PLA), Beijing, China
| | - Jinlong Dai
- Department of Pathology, The Eighth Medical Center, General Hospital of the Chinese People's Liberation Army (PLA), Beijing, China
| | - Yan Yang
- Department of Radiology, The Eighth Medical Center, General Hospital of the Chinese People's Liberation Army (PLA), Beijing, China
| | - Yinghui Chai
- Department of Clinical Laboratory, The Eighth Medical Center, General Hospital of the Chinese People's Liberation Army (PLA), Beijing, China
| | - Wen Chen
- Department of Pathology, The Eighth Medical Center, General Hospital of the Chinese People's Liberation Army (PLA), Beijing, China
| |
Collapse
|
4
|
Najah Q, Almosilhy NA, Ghanm TIE. Is nivolumab alone or in combination with ipilimumab more effective for treating lung cancer? a meta-analysis. Eur J Clin Pharmacol 2025; 81:269-278. [PMID: 39680077 DOI: 10.1007/s00228-024-03789-0] [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: 07/08/2024] [Accepted: 12/08/2024] [Indexed: 12/17/2024]
Abstract
BACKGROUND Nivolumab and ipilimumab combination immunotherapy has become a standard treatment option for certain cancers. However, the benefits of combination therapy compared to nivolumab monotherapy in lung cancer patients are not entirely clear. We aimed to evaluate whether nivolumab plus ipilimumab improves clinical outcomes in lung cancer patients compared to nivolumab monotherapy. METHODS A literature search was performed on PubMed, Web of Science, and Scopus from inception until November 2024 to identify relevant randomized controlled trials. The Cochrane risk of bias tool was used to assess the risk of bias, the hazard ratio (HR) was calculated for survival, risk ratios (RRs) were calculated for response rate and safety outcomes, and a random effects model meta-analysis was performed to estimate the safety and efficacy of the treatments. RESULTS Seven trials comprising 2134 patients were included. Compared with patients receiving nivolumab monotherapy, non-small cell lung cancer patients who received combination therapy had better progression-free survival (HR = 0.82, 95% CI 0.71; 0.93, P < 0.01, low certainty), and there were no significant differences in overall survival (HR = 0.95, 95% CI 0.86; 1.0, P = 0.31, moderate certainty), or objective response rate (RR = 1.36, 95% CI 0.91; 2.02, P = 0.14 very low certainty). The combination group had a significantly greater risk of grade 3-4 adverse events (RR = 2.77, 95% CI 1.38; 5.56, P < 0.01, low certainty). CONCLUSION Although combination treatment significantly improved progression-free survival in NSCLC patients, it was also associated with a greater risk of adverse events and treatment-related mortality than nivolumab monotherapy. The current evidence is insufficient for choosing combination treatment over nivolumab monotherapy.
Collapse
Affiliation(s)
- Qasi Najah
- Faculty of Medicine, Elmergib University, Al-Khums, Libya.
- Medical Research Group of Libya, Negida Academy, Arlington, MA, USA.
| | - Nereen A Almosilhy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta, Egypt
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
| | - Thoria Ibrahim Essa Ghanm
- Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Medical Research Group of Egypt, Negida Academy, Arlington, MA, USA
| |
Collapse
|
5
|
Cuenot L, Valnet-Rabier MB, Bendjama A, Aubin F, Fischer S, Viot J, Nerich V. [Serious adverse effects with immunotherapies for the treatment of melanoma, non-small cell lung cancer, and renal cell carcinoma: Real-world evidence study]. Bull Cancer 2024; 111:1111-1121. [PMID: 39389873 DOI: 10.1016/j.bulcan.2024.08.017] [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: 04/29/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 10/12/2024]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) are a key component of standard anticancer systemic therapy. While their immune-related adverse effects (irAEs) have been widely described, there are few data on grade≥3 irAEs. The primary aim of our descriptive study was to evaluate their incidence and characteristics. METHODS An observational, retrospective, monocentric study was conducted. It included patients with locally advanced or metastatic melanoma, non-small cell lung cancer or renal cell carcinoma who initiated ICI therapy between 2016-2021 and experienced at least one grade≥3 irAEs coded according to the MedDRA® system. RESULTS All cancer types and ICIs combined, the incidence of grade≥3 irAEs was estimated at 11.7% [9.6-13.9]. These were mainly hepatobiliary (22%), gastrointestinal (17%), musculoskeletal (16%) and respiratory (16%) disorders. They occurred on average 6.2±6.2 months after the start of treatment, resulting in hospitalization or prolonged hospitalization in over 40 and 20% of cases, respectively. Resolution without sequelae was observed in 56% of cases, but four patients died. DISCUSSION This real-world study investigated three cancers and several ICIs, unlike previously published studies that focused on a single cancer and/or one ICI. It provides a better understanding of grade≥3 irAEs, most of which are reversible, which an aim to optimize patient care.
Collapse
Affiliation(s)
- Léa Cuenot
- Pôle pharmacie, CHU de Besançon, 25030 Besançon, France
| | | | | | - François Aubin
- Service de dermatologie, Inserm, EFS-BFC, UMR 1098, CHU de Besançon, université de Franche-Comté, 25030 Besançon, France
| | - Sarah Fischer
- Pôle pharmacie, CHU de Besançon, 25030 Besançon, France
| | - Julien Viot
- Service de pneumologie, Inserm, EFS-BFC, UMR 1098, CHU de Besançon, université de Franche-Comté, 25030 Besançon, France
| | - Virginie Nerich
- Pôle pharmacie, Inserm, EFS-BFC, UMR 1098, CHU de Besançon, université de Franche-Comté, 25030 Besançon, France.
| |
Collapse
|
6
|
Boscolo Bragadin A, Del Bianco P, Zulato E, Attili I, Pavan A, Carlet J, Marra L, Guarneri V, Indraccolo S, Bonanno L. Longitudinal liquid biopsy predicts clinical benefit from immunotherapy in advanced non-small cell lung cancer. NPJ Precis Oncol 2024; 8:234. [PMID: 39420036 PMCID: PMC11486993 DOI: 10.1038/s41698-024-00704-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 09/11/2024] [Indexed: 10/19/2024] Open
Abstract
High heterogeneity in clinical benefit characterizes the use of immune checkpoint inhibitors (ICIs) in non-small cell lung cancer (NSCLC). We prospectively enrolled 113 advanced NSCLC patients treated with ICIs and performed liquid biopsy at the time of ICI start (T1), after 3 weeks (T2) and at the time of radiological evaluation (T3). Molecular variables were associated with outcome endpoints: cfDNA quantification, its dynamic change (∆T2-T1), variant allele frequency (VAF) of the gene with the highest frequency detected at baseline with NGS (maxVAF) and its dynamic change (∆T2-T1). At multivariate analysis, PD-L1 negativity, T1 cfDNA, cfDNA increase (∆T2-T1), and T2 VAF were significantly associated with shorter progression-free survival (PFS); PD-L1 negativity, squamous histology, T1 cfDNA, cfDNA (∆T2-T1) increase, and T2 maxVAF affected overall survival (OS). Among high PD-L1 expressing patients treated in first-line, elevated T2 maxVAF and cfDNA increase (∆T2-T1) correlated with worse PFS; higher T2 maxVAF and cfDNA increase (∆T2-T1) with worse OS. Derived integrated models were used to build nomograms and categorize different risk groups.
Collapse
Affiliation(s)
- Andrea Boscolo Bragadin
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Paola Del Bianco
- Clinical Research Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Elisabetta Zulato
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Ilaria Attili
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Alberto Pavan
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Jessica Carlet
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Ludovica Marra
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
| | - Stefano Indraccolo
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Laura Bonanno
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy.
- Medical Oncology 2, Veneto Institute of Oncology IOV - IRCCS, Padua, Italy.
| |
Collapse
|
7
|
Matsumoto H, Fukushima T, Kobayashi N, Higashino Y, Muraoka S, Ohtsu Y, Hirata M, Somekawa K, Kaneko A, Nagasawa R, Kubo S, Tanaka K, Murohashi K, Fujii H, Watanabe K, Horita N, Hara Y, Kaneko T. High red blood cell distribution width attenuates the effectiveness of Immune checkpoint inhibitor therapy: An exploratory study using a clinical data warehouse. PLoS One 2024; 19:e0299760. [PMID: 39088539 PMCID: PMC11293676 DOI: 10.1371/journal.pone.0299760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/15/2024] [Indexed: 08/03/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have improved outcomes in cancer treatment but are also associated with adverse events and financial burdens. Identifying accurate biomarkers is crucial for determining which patients are likely to benefit from ICIs. Current markers, such as PD-L1 expression and tumor mutation burden, exhibit limited predictive accuracy. This study utilizes a Clinical Data Warehouse (CDW) to explore the prognostic significance of novel blood-based factors, such as the neutrophil-to-lymphocyte ratio and red cell distribution width (RDW), to enhance the prediction of ICI therapy benefit. METHODS This retrospective study utilized an exploratory cohort from the CDW that included a variety of cancers to explore factors associated with pembrolizumab treatment duration, validated in a non-small cell lung cancer (NSCLC) patient cohort from electronic medical records (EMR) and CDW. The CDW contained anonymized data on demographics, diagnoses, medications, and tests for cancer patients treated with ICIs between 2017-2022. Logistic regression identified factors predicting ≤2 or ≥5 pembrolizumab doses as proxies for progression-free survival (PFS), and Receiver Operating Characteristic analysis was used to examine their predictive ability. These factors were validated by correlating doses with PFS in the EMR cohort and re-testing their significance in the CDW cohort with other ICIs. This dual approach utilized the CDW for discovery and EMR/CDW cohorts for validating prognostic biomarkers before ICI treatment. RESULTS A total of 609 cases (428 in the exploratory cohort and 181 in the validation cohort) from CDW and 44 cases from EMR were selected for study. CDW analysis revealed that elevated red cell distribution width (RDW) correlated with receiving ≤2 pembrolizumab doses (p = 0.0008), with an AUC of 0.60 for predicting treatment duration. RDW's correlation with PFS (r = 0.80, p<0.0001) and its weak association with RDW (r = -0.30, p = 0.049) were confirmed in the EMR cohort. RDW also remained significant in predicting short treatment duration across various ICIs (p = 0.0081). This dual methodology verified pretreatment RDW elevation as a prognostic biomarker for shortened ICI therapy. CONCLUSION This study suggests the utility of CDWs in identifying prognostic biomarkers for ICI therapy in cancer treatment. Elevated RDW before treatment initiation emerged as a potential biomarker of shorter therapy duration.
Collapse
Affiliation(s)
- Hiromi Matsumoto
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | | | - Nobuaki Kobayashi
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yuuki Higashino
- Yokohama City University School of Medicine, Yokohama, Japan
| | - Suguru Muraoka
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yukiko Ohtsu
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Momo Hirata
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kohei Somekawa
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ayami Kaneko
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Ryo Nagasawa
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Sousuke Kubo
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Katsushi Tanaka
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Kota Murohashi
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Hiroaki Fujii
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Keisuke Watanabe
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Nobuyuki Horita
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Yu Hara
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| | - Takeshi Kaneko
- Department of Pulmonology, Yokohama City University Graduate School of Medicine, Yokohama, Japan
| |
Collapse
|
8
|
Tokito T, Kolesnik O, Sørensen J, Artac M, Quintela ML, Lee JS, Hussein M, Pless M, Paz-Ares L, Leopold L, Daniel J, Munteanu M, Samkari A, Xu L, Butts C. Epacadostat plus pembrolizumab versus placebo plus pembrolizumab as first-line treatment for metastatic non-small cell lung cancer with high levels of programmed death-ligand 1: a randomized, double-blind phase 2 study. BMC Cancer 2024; 23:1251. [PMID: 39054476 PMCID: PMC11270761 DOI: 10.1186/s12885-023-11203-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 07/19/2023] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Pembrolizumab is a first-line therapy for certain patients with advanced/metastatic non-small cell lung cancer (NSCLC). Combining pembrolizumab with other immunotherapies may enhance tumor cell killing and clinical outcomes. Epacadostat is a selective inhibitor of indoleamine 2,3-dioxygenase 1, an immuno-regulatory enzyme involved in tryptophan to kynurenine metabolism that inhibits T cell-mediated immune responses. METHODS In this randomized phase II study, patients with metastatic NSCLC expressing high (≥ 50%) programmed death-ligand 1 (PD-L1) levels received pembrolizumab 200 mg every 21 days plus oral epacadostat 100 mg twice daily (combination) or matching placebo (control). The primary objective was objective response rate (ORR); secondary objectives were progression-free survival (PFS), overall survival (OS), duration of response (DOR) and safety/tolerability. RESULTS 154 patients were randomized (77 per group). Median (range) follow-up was 6.8 months (0.1-11.4) and 7.0 months (0.2-11.9) in the combination and control groups, respectively Confirmed ORR was similar between groups (combination: 32.5%, 95% CI 22.2-44.1; control: 39.0%, 95% CI 28.0-50.8; difference: - 6.5, 95% CI - 21.5 to 8.7; 1-sided P = 0.8000). Median (range) DOR was 6.2 months (1.9 + to 6.5 +) and not reached (1.9 + to 8.6 +) in the combination and control groups, respectively. Although not formally tested, median PFS was 6.7 and 6.2 months for the combination and control groups, respectively, and median OS was not reached in either group. Circulating kynurenine levels increased from C1D1 to C2D1 (P < 0.01) in the control group and decreased from C1D1 to C2D1 (P < 0.01) in the combination group but were not normalized in most patients. The most frequent serious adverse events (AEs) (≥ 2%) were pneumonia (4.0%), anemia (2.7%), atelectasis (2.7%) and pneumonitis (2.7%) in the combination group and pneumonia (3.9%), pneumonitis (2.6%) and hypotension (2.6%) in the control group. Two deaths due to drug-related AEs were reported, both in the control group. CONCLUSIONS Addition of epacadostat to pembrolizumab therapy for PD-L1-high metastatic NSCLC was generally well tolerated but did not demonstrate an improved therapeutic effect. Evaluating higher doses of epacadostat that normalize kynurenine levels when given in combination with checkpoint inhibitors may be warranted. TRIAL REGISTRATION ClinicalTrials.gov, NCT03322540. Registered 10/26/2017.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Female
- Humans
- Male
- Middle Aged
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- B7-H1 Antigen/antagonists & inhibitors
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/mortality
- Double-Blind Method
- Lung Neoplasms/drug therapy
- Lung Neoplasms/pathology
- Lung Neoplasms/mortality
- Oximes/administration & dosage
- Oximes/therapeutic use
- Oximes/adverse effects
- Progression-Free Survival
- Sulfonamides/administration & dosage
- Sulfonamides/therapeutic use
- Sulfonamides/adverse effects
- Neoplasm Metastasis
- Placebos
Collapse
Affiliation(s)
- Takaaki Tokito
- Division of Respirology, Neurology and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830-0011, Japan.
| | - Oleksii Kolesnik
- Zaporizhzhya Regional Clinical Oncology Center, Zaporizhzhya State Medical University, Zaporozhye, Ukraine
| | - Jens Sørensen
- Department of Oncology, Centre for Cancer and Organ Diseases, Rigshospitalet, Copenhagen, Denmark
| | - Mehmet Artac
- Department of Medical Oncology, Necmettin Erbakan Universitesi Meram Tip Fakultesi Hastanesi, Konya, Turkey
| | - Martín Lázaro Quintela
- Department of Medical Oncology, Hospital Álvaro Cunqueiro-Complexo Hospitalario Universitario de Vigo, Vigo, Spain
| | - Jong-Seok Lee
- Division of Hematology-Oncology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | | | - Miklos Pless
- Department of Medical Oncology, Kantonsspital Winterthur, Winterthur, Switzerland
| | - Luis Paz-Ares
- Department of Medical Oncology, Hospital Universitario, 12 de Octubre, Universidad Complutense & Ciberonc, Madrid, Spain
| | | | | | | | | | - Lu Xu
- Merck & Co., Inc., Rahway, NJ, United States
| | - Charles Butts
- Department of Oncology, Division of Medical Oncology, Cross Cancer Institute, Edmonton, Canada
| |
Collapse
|
9
|
Chatelain C, Berland L, Grard M, Jouand N, Fresquet J, Nader J, Hirigoyen U, Petithomme T, Combredet C, Pons-Tostivint E, Fradin D, Treps L, Blanquart C, Boisgerault N, Tangy F, Fonteneau JF. Interplay between oncolytic measles virus, macrophages and cancer cells induces a proinflammatory tumor microenvironment. Oncoimmunology 2024; 13:2377830. [PMID: 39005546 PMCID: PMC11244337 DOI: 10.1080/2162402x.2024.2377830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024] Open
Abstract
Attenuated measles virus (MV) exerts its oncolytic activity in malignant pleural mesothelioma (MPM) cells that lack type-I interferon (IFN-I) production or responsiveness. However, other cells in the tumor microenvironment (TME), such as myeloid cells, possess functional antiviral pathways. In this study, we aimed to characterize the interplay between MV and the myeloid cells in human MPM. We cocultured MPM cell lines with monocytes or macrophages and infected them with MV. We analyzed the transcriptome of each cell type and studied their secretion and phenotypes by high-dimensional flow cytometry. We also measured transgene expression using an MV encoding GFP (MV-GFP). We show that MPM cells drive the differentiation of monocytes into M2-like macrophages. These macrophages inhibit GFP expression in tumor cells harboring a defect in IFN-I production and a functional signaling downstream of the IFN-I receptor, while having minimal effects on GFP expression in tumor cells with defect of responsiveness to IFN-I. Interestingly, inhibition of the IFN-I signaling by ruxolitinib restores GFP expression in tumor cells. Upon MV infection, cocultured macrophages express antiviral pro-inflammatory genes and induce the expression of IFN-stimulated genes in tumor cells. MV also increases the expression of HLA and costimulatory molecules on macrophages and their phagocytic activity. Finally, MV induces the secretion of inflammatory cytokines, especially IFN-I, and PD-L1 expression in tumor cells and macrophages. These results show that macrophages reduce viral proteins expression in some MPM cell lines through their IFN-I production and generate a pro-inflammatory interplay that may stimulate the patient's anti-tumor immune response.
Collapse
Affiliation(s)
- Camille Chatelain
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Laurine Berland
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Marion Grard
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Nicolas Jouand
- LabEx IGO, Nantes Université, Nantes, France
- Nantes Université, CHU Nantes, CNRS, Inserm, BioCore, US16, SFR Bonamy, Nantes, France
| | - Judith Fresquet
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Joëlle Nader
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Ugo Hirigoyen
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Tacien Petithomme
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Chantal Combredet
- Vaccines Innovation Laboratory, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Elvire Pons-Tostivint
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
- Centre Hospitalier Universitaire Nantes, Medical Oncology, Nantes University, Nantes, France
| | - Delphine Fradin
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Lucas Treps
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Christophe Blanquart
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Nicolas Boisgerault
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| | - Frédéric Tangy
- Vaccines Innovation Laboratory, Institut Pasteur, Université de Paris Cité, Paris, France
- Oncovita, Paris, France
| | - Jean-François Fonteneau
- Nantes Université, Inserm UMR 1307, CNRS UMR 6075, Université d’Angers, Nantes, France
- LabEx IGO, Nantes Université, Nantes, France
| |
Collapse
|
10
|
Besse B, Pons-Tostivint E, Park K, Hartl S, Forde PM, Hochmair MJ, Awad MM, Thomas M, Goss G, Wheatley-Price P, Shepherd FA, Florescu M, Cheema P, Chu QSC, Kim SW, Morgensztern D, Johnson ML, Cousin S, Kim DW, Moskovitz MT, Vicente D, Aronson B, Hobson R, Ambrose HJ, Khosla S, Reddy A, Russell DL, Keddar MR, Conway JP, Barrett JC, Dean E, Kumar R, Dressman M, Jewsbury PJ, Iyer S, Barry ST, Cosaert J, Heymach JV. Biomarker-directed targeted therapy plus durvalumab in advanced non-small-cell lung cancer: a phase 2 umbrella trial. Nat Med 2024; 30:716-729. [PMID: 38351187 PMCID: PMC10957481 DOI: 10.1038/s41591-024-02808-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 01/09/2024] [Indexed: 03/23/2024]
Abstract
For patients with non-small-cell lung cancer (NSCLC) tumors without currently targetable molecular alterations, standard-of-care treatment is immunotherapy with anti-PD-(L)1 checkpoint inhibitors, alone or with platinum-doublet therapy. However, not all patients derive durable benefit and resistance to immune checkpoint blockade is common. Understanding mechanisms of resistance-which can include defects in DNA damage response and repair pathways, alterations or functional mutations in STK11/LKB1, alterations in antigen-presentation pathways, and immunosuppressive cellular subsets within the tumor microenvironment-and developing effective therapies to overcome them, remains an unmet need. Here the phase 2 umbrella HUDSON study evaluated rational combination regimens for advanced NSCLC following failure of anti-PD-(L)1-containing immunotherapy and platinum-doublet therapy. A total of 268 patients received durvalumab (anti-PD-L1 monoclonal antibody)-ceralasertib (ATR kinase inhibitor), durvalumab-olaparib (PARP inhibitor), durvalumab-danvatirsen (STAT3 antisense oligonucleotide) or durvalumab-oleclumab (anti-CD73 monoclonal antibody). Greatest clinical benefit was observed with durvalumab-ceralasertib; objective response rate (primary outcome) was 13.9% (11/79) versus 2.6% (5/189) with other regimens, pooled, median progression-free survival (secondary outcome) was 5.8 (80% confidence interval 4.6-7.4) versus 2.7 (1.8-2.8) months, and median overall survival (secondary outcome) was 17.4 (14.1-20.3) versus 9.4 (7.5-10.6) months. Benefit with durvalumab-ceralasertib was consistent across known immunotherapy-refractory subgroups. In ATM-altered patients hypothesized to harbor vulnerability to ATR inhibition, objective response rate was 26.1% (6/23) and median progression-free survival/median overall survival were 8.4/22.8 months. Durvalumab-ceralasertib safety/tolerability profile was manageable. Biomarker analyses suggested that anti-PD-L1/ATR inhibition induced immune changes that reinvigorated antitumor immunity. Durvalumab-ceralasertib is under further investigation in immunotherapy-refractory NSCLC.ClinicalTrials.gov identifier: NCT03334617.
Collapse
Affiliation(s)
- Benjamin Besse
- Institut Gustave Roussy, Paris-Saclay University, Villejuif, France
| | - Elvire Pons-Tostivint
- Medical Oncology, Centre Hospitalier Universitaire Nantes, Nantes University, Nantes, France
| | - Keunchil Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
- MD Anderson Cancer Center, Houston, TX, USA
| | - Sylvia Hartl
- Ludwig Boltzmann Institute for Lung Health, Clinic Penzing, Vienna, Austria
- Sigmund Freud University, Vienna, Austria
| | - Patrick M Forde
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Maximilian J Hochmair
- Department of Respiratory and Critical Care Medicine, Karl Landsteiner Institute of Lung Research and Pulmonary Oncology, Klinik Floridsdorf, Vienna, Austria
| | - Mark M Awad
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michael Thomas
- Department of Thoracic Oncology, Thoraxklinik, Heidelberg University Hospital and National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Germany; Translational Lung Research Center Heidelberg (TLRC-H), Member of the German Center for Lung Research (DZL), Heidelberg, Germany
| | - Glenwood Goss
- The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Paul Wheatley-Price
- The Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - Frances A Shepherd
- Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Marie Florescu
- Division of Hematology Oncology, Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - Parneet Cheema
- William Osler Health System, University of Toronto, Toronto, Ontario, Canada
| | | | - Sang-We Kim
- Department of Oncology, Asan Medical Center, Seoul, Republic of Korea
| | - Daniel Morgensztern
- Department of Medicine, Division of Medical Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Melissa L Johnson
- Sarah Cannon Research Institute, Tennessee Oncology, Nashville, TN, USA
| | - Sophie Cousin
- Department of Medical Oncology, Institut Bergonié, Regional Comprehensive Cancer Center, Bordeaux, France
| | - Dong-Wan Kim
- Seoul National University College of Medicine and Seoul National University Hospital, Seoul, Republic of Korea
| | - Mor T Moskovitz
- Institute of Oncology, Rambam Medical Center, Haifa, Israel
- Thoracic Cancer Service, Rabin Medical Center Davidoff Cancer Centre, Beilinson Campus, Petah Tikva, Israel
| | - David Vicente
- Department of Medical Oncology, Hospital Universitario Virgen Macarena, Seville, Spain
| | - Boaz Aronson
- Oncology Early Global Development, AstraZeneca, Gaithersburg, MD, USA
| | | | - Helen J Ambrose
- Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Sajan Khosla
- Real-World Evidence, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Avinash Reddy
- Oncology Data Science, Oncology R&D, AstraZeneca, Boston, MA, USA
| | - Deanna L Russell
- Translational Medicine, Oncology R&D, AstraZeneca, Boston, MA, USA
| | - Mohamed Reda Keddar
- Oncology Data Science, Research and Early Development, Oncology R&D, AstraZeneca, Cambridge, UK
| | - James P Conway
- Oncology Data Science, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - J Carl Barrett
- Translational Medicine, Oncology R&D, AstraZeneca, Boston, MA, USA
| | - Emma Dean
- Oncology R&D, AstraZeneca, Cambridge, UK
| | - Rakesh Kumar
- Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | | | | | - Sonia Iyer
- Translational Medicine, Oncology R&D, AstraZeneca, Boston, MA, USA
| | | | | | - John V Heymach
- Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
11
|
de Miguel-Perez D, Pickering EM, Malapelle U, Grier W, Pepe F, Pisapia P, Russo G, Pinto JA, Russo A, Troncone G, Culligan MJ, Scilla KA, Mehra R, Mohindra P, Arrieta O, Cardona AF, Del Re M, Sachdeva A, Hirsch FR, Wolf A, Friedberg JS, Rolfo C. Genomic profiling of tissue and blood predicts survival outcomes in patients with resected pleural mesothelioma. Eur J Cancer 2024; 196:113457. [PMID: 38008032 DOI: 10.1016/j.ejca.2023.113457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 11/28/2023]
Abstract
PURPOSE Pleural mesothelioma (PM) is an aggressive tumor still considered incurable, in part due to the lack of predictive biomarkers. Little is known about the clinical implications of molecular alterations in resectable PM tissues and blood. Here, we characterized genetic alterations to identify prognostic and predictive biomarkers in patients with resected PM. EXPERIMENTAL DESIGN Targeted next-generation sequencing was performed in retrospective pleural tumor tissue and paired plasma samples from stage IB-IIIB resected PM. Association between prognosis and presence of specific mutations was validated in silico. RESULTS Thirty PM tissues and paired blood samples from 12 patients were analyzed. High tissue tumor mutational burden (TMB) (>10 mutations/Mb), tissue median minor allele frequency (MAF) (>9 mutations/Mb), and blood TMB (>6 mutations/Mb), tissue KMT2C, PBRM1, PKHD1,EPHB1 and blood LIFR mutations correlated with longer disease-free survival and/or overall survival. High concordance (>80%) between tissue and blood was found for some mutations. CONCLUSIONS Tissue TMB and MAF, blood TMB, and specific mutations correlated with outcomes in patients with resected PM and should be further studied to validate their role as prognostic biomarkers and potentially predictive factors for combinations with immune-checkpoint inhibitors. This suggest that molecular profiling could identify longer survivors in patients with resected PM.
Collapse
Affiliation(s)
- Diego de Miguel-Perez
- Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Edward M Pickering
- Section of Interventional Pulmonology, Division of Pulmonary and Critical Care Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - William Grier
- Division of Pulmonary and Critical Care Medicine, University of Maryland Medical Center, Baltimore, MD, USA
| | - Francesco Pepe
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Pasquale Pisapia
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Gianluca Russo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Joseph A Pinto
- Centro de Investigación Básica y Traslacional, Auna Ideas, Lima, Peru
| | - Alessandro Russo
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Giancarlo Troncone
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Melissa J Culligan
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA, USA
| | - Katherine A Scilla
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Ranee Mehra
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Pranshu Mohindra
- Department of Radiation Oncology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Oscar Arrieta
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Andres F Cardona
- Luis Carlos Sarmiento Angulo Cancer Treatment and Research Center (CTIC) / Foundation for Clinical and Applied Cancer Research (FICMAC) / Molecular Oncology and Biology Systems Research Group (Fox-G), Universidad El Bosque, Bogotá, Colombia
| | - Marzia Del Re
- Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ashutosh Sachdeva
- Section of Interventional Pulmonology, Division of Pulmonary and Critical Care Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Fred R Hirsch
- Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrea Wolf
- Department of Thoracic Surgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Joseph S Friedberg
- Department of Thoracic Medicine and Surgery, Temple University, Philadelphia, PA, USA
| | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
12
|
Hikmet F, Rassy M, Backman M, Méar L, Mattsson JSM, Djureinovic D, Botling J, Brunnström H, Micke P, Lindskog C. Expression of cancer-testis antigens in the immune microenvironment of non-small cell lung cancer. Mol Oncol 2023; 17:2603-2617. [PMID: 37341056 DOI: 10.1002/1878-0261.13474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 05/15/2023] [Accepted: 06/19/2023] [Indexed: 06/22/2023] Open
Abstract
The antigenic repertoire of tumors is critical for successful anti-cancer immune response and the efficacy of immunotherapy. Cancer-testis antigens (CTAs) are targets of humoral and cellular immune reactions. We aimed to characterize CTA expression in non-small cell lung cancer (NSCLC) in the context of the immune microenvironment. Of 90 CTAs validated by RNA sequencing, eight CTAs (DPEP3, EZHIP, MAGEA4, MAGEB2, MAGEC2, PAGE1, PRAME, and TKTL1) were selected for immunohistochemical profiling in cancer tissues from 328 NSCLC patients. CTA expression was compared with immune cell densities in the tumor environment and with genomic, transcriptomic, and clinical data. Most NSCLC cases (79%) expressed at least one of the analyzed CTAs, and CTA protein expression correlated generally with RNA expression. CTA profiles were associated with immune profiles: high MAGEA4 expression was related to M2 macrophages (CD163) and regulatory T cells (FOXP3), low MAGEA4 was associated with T cells (CD3), and high EZHIP was associated with plasma cell infiltration (adj. P-value < 0.05). None of the CTAs correlated with clinical outcomes. The current study provides a comprehensive evaluation of CTAs and suggests that their association with immune cells may indicate in situ immunogenic effects. The findings support the rationale to harness CTAs as targets for immunotherapy.
Collapse
Affiliation(s)
- Feria Hikmet
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden
| | - Marc Rassy
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden
| | - Max Backman
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden
| | - Loren Méar
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden
| | | | - Dijana Djureinovic
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden
- Department of Medicine (Medical Oncology), Yale University School of Medicine, New Haven, CT, USA
| | - Johan Botling
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden
| | - Hans Brunnström
- Division of Pathology, Department of Clinical Sciences Lund, Lund University, Sweden
| | - Patrick Micke
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden
| | - Cecilia Lindskog
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Sweden
| |
Collapse
|
13
|
Goldschmidt JH, Chou L, Chan PK, Chen L, Robert N, Kinsey J, Pitts K, Nestor M, Rock EP, Lazarus HM. Real-world outcomes of 18,186 metastatic solid tumor outpatients: Baseline blood cell counts correlate with survival after immune checkpoint inhibitor therapy. Cancer Med 2023; 12:20783-20797. [PMID: 37962239 PMCID: PMC10709745 DOI: 10.1002/cam4.6645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 09/27/2023] [Accepted: 10/16/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Patient survival in advanced/metastatic melanoma, non-small cell lung cancer (NSCLC), and renal cell carcinoma (RCC) has improved with immune checkpoint inhibitors (ICI). Biomarkers' role in prognosis and treatment has been limited by conflicting trial results. METHODS This retrospective, observational study analyzed baseline demographic, clinical, laboratory, and treatment data versus outcomes of The US Oncology Network adult outpatients. Patients with advanced/metastatic melanoma, NSCLC, or RCC treated between January 1, 2015 and November 30, 2020 were given ICI monotherapy or combination therapy with ipilimumab, pembrolizumab, nivolumab, or atezolizumab. Treatment outcomes (overall survival [OS], time to treatment discontinuation, time to next treatment) were followed longitudinally until May 31, 2021, last patient record, or date of death. Baseline blood cell counts, including absolute monocyte count (AMC), absolute lymphocyte count (ALC), monocyte-to-lymphocyte ratio (MLR), absolute neutrophil count (ANC), and eosinophil count, were subdivided into quintiles for univariate and multivariable Cox regression analyses. RESULTS Data from 18,186 patients with advanced/metastatic melanoma (n = 3314), NSCLC (n = 12,416), and RCC (n = 2456) were analyzed. Better OS correlated with increased baseline serum albumin concentration, increased eosinophil and lymphocyte counts, and Western United States physician practice location. Decreased OS correlated with increased AMC, MLR, ANC, age, and worse Eastern Cooperative Oncology Group performance status. CONCLUSIONS To our knowledge, this study is the largest to date to associate baseline survival indicators and outcomes in outpatients with advanced/metastatic melanoma, NSCLC, or RCC and receiving ICIs. Results may inform disease-specific prognostic models and help providers identify patients most likely to benefit from ICI therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Joyce Kinsey
- Partner Therapeutics, IncLexingtonMassachusettsUSA
| | | | - Matt Nestor
- Partner Therapeutics, IncLexingtonMassachusettsUSA
| | | | - Hillard M. Lazarus
- Department of Medicine, Division of Hematology and OncologyCase Western Reserve UniversityClevelandOhioUSA
| |
Collapse
|
14
|
Chen R, Shao C, Liu X, Huang H, Pan B, Xu K, Zhu R, Li M, Zhao Y, Chen K, Wang M, Xu Z. Clinical spectrum of Chinese hospitalized lung cancer patients with concomitant interstitial lung disease: before and after the new era of LC treatment. Clin Exp Med 2023; 23:2321-2330. [PMID: 36715832 PMCID: PMC9885922 DOI: 10.1007/s10238-023-00999-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/16/2023] [Indexed: 01/31/2023]
Abstract
This study aimed to explore the general characteristics and spectrum of hospitalized Chinese patients suffering from lung cancer with concomitant interstitial lung disease (LC-ILD). Furthermore, we compared their features before and after the period of immunotherapy for lung cancer. A retrospective analysis of the clinical characteristics of hospitalized LC patients with definite pathological diagnoses was performed from 2014 to 2021. ILD was defined after the review of chest CT imaging. There were 13,085 hospitalized LC patients. Among them, 509 patients (3.89%) had 551 cases of ILD. There were variable underlying causes of ILD, including idiopathic interstitial pneumonia (360 patients), LC treatment-associated ILD (134 cases), and connective tissue disease-associated ILD (55 patients). Although most LC-ILD patients were suffering from adenocarcinoma (204/40.1%), SCLC patients were prone to concomitant ILD (10.8% of all SCLC cases), followed by SCC (9.6% of all SCC cases). All but 10 LC-ILD patients received anti-LC treatment; however, only 39 (10.8%) LC-IIP patients received anti-ILD treatment. There were more LC-ILD patients in the 2018-2021 group than in the 2014-2017 group (5.16% vs. 2.03%, p < 0.001). The underlying causes of ILD were significantly different between the 2018-2021 group and the 2014-2017 group (p < 0.001). After adjusting for the number of hospitalized patients having the same LC pathological pattern, SCLC was determined to be the most likely to be concomitant with ILD, followed by SCC. Most LC-ILD patients were scheduled for anti-LC therapy; however, treatments for concomitant IIP were usually ignored. LC treatment-associated ILD should receive more attention than before.
Collapse
Affiliation(s)
- Ruxuan Chen
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730 China
| | - Chi Shao
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730 China
| | - Xiangning Liu
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730 China
| | - Hui Huang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730 China
| | - Boju Pan
- Pathological Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730 China
| | - Kai Xu
- Radiological Department, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730 China
| | - Rui Zhu
- Medical Records Department, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730 China
| | - Mei Li
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730 China
| | - Yang Zhao
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730 China
| | - Keqi Chen
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730 China
| | - Mengzhao Wang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730 China
| | - Zuojun Xu
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No.1 Shuaifuyuan Street, Dongcheng District, Beijing, 100730 China
| |
Collapse
|
15
|
Foy V, McNamara MG, Valle JW, Lamarca A, Edeline J, Hubner RA. Current Evidence for Immune Checkpoint Inhibition in Advanced Hepatocellular Carcinoma. Curr Oncol 2023; 30:8665-8685. [PMID: 37754543 PMCID: PMC10529518 DOI: 10.3390/curroncol30090628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/11/2023] [Accepted: 09/18/2023] [Indexed: 09/28/2023] Open
Abstract
The treatment of advanced unresectable HCC (aHCC) remains a clinical challenge, with limited therapeutic options and poor prognosis. The results of IMbrave150 and HIMALAYA have changed the treatment paradigm for HCC and established immune checkpoint inhibition (ICI), either combined with anti-angiogenic therapy or dual ICI, as preferred first-line therapy for eligible patients with aHCC. Numerous other combination regimens involving ICI are under investigation with the aim of improving the tumour response and survival of patients with all stages of HCC. This review will explore the current evidence for ICI in patients with advanced HCC and discuss future directions, including the unmet clinical need for predictive biomarkers to facilitate patient selection, the effects of cirrhosis aetiology on response to ICI, and the safety of its use in patients with impaired liver function.
Collapse
Affiliation(s)
- Victoria Foy
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Rd., Manchester M20 4BX, UK
| | - Mairéad G. McNamara
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Rd., Manchester M20 4BX, UK
- Division of Cancer Sciences, University of Manchester, Oxford Rd., Manchester M13 9PL, UK
| | - Juan W. Valle
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Rd., Manchester M20 4BX, UK
- Division of Cancer Sciences, University of Manchester, Oxford Rd., Manchester M13 9PL, UK
| | - Angela Lamarca
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Rd., Manchester M20 4BX, UK
- Division of Cancer Sciences, University of Manchester, Oxford Rd., Manchester M13 9PL, UK
- Department of Oncology, OncoHealth Institute, Fundación Jiménez Díaz University Hospital, Avenida de los Reyes Catolicos 2, 28040 Madrid, Spain
| | - Julien Edeline
- Centre Eugène Marquis, Av. de la Bataille Flandres Dunkerque-CS 44229, CEDEX, 35042 Rennes, France;
| | - Richard A. Hubner
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Rd., Manchester M20 4BX, UK
- Division of Cancer Sciences, University of Manchester, Oxford Rd., Manchester M13 9PL, UK
| |
Collapse
|
16
|
Deboever N, Zhou N, McGrail DJ, Tomczak K, Oliva JL, Feldman HA, Parra E, Zhang J, Lee PP, Antonoff MB, Hofstetter WL, Mehran RJ, Rajaram R, Rice DC, Roth JA, Swisher SS, Vaporciyan AA, Altan M, Weissferdt A, Tsao AS, Haymaker CL, Sepesi B. Radiographic response to neoadjuvant therapy in pleural mesothelioma should serve as a guide for patient selection for cytoreductive operations. Front Oncol 2023; 13:1216999. [PMID: 37637041 PMCID: PMC10455934 DOI: 10.3389/fonc.2023.1216999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 07/10/2023] [Indexed: 08/29/2023] Open
Abstract
Background Malignant pleural mesothelioma (MPM) is associated with poor prognosis despite advances in multimodal therapeutic strategies. While patients with resectable disease may benefit from added survival with oncologic resection, patient selection for mesothelioma operations often relies on both objective and subjective evaluation metrics. We sought to evaluate factors associated with improved overall survival (OS) in patients with mesothelioma who underwent macroscopic complete resection (MCR). Methods Patients with MPM who received neoadjuvant therapy and underwent MCR were identified in a prospectively maintained departmental database. Clinicopathologic, blood-based, and radiographic variables were collected and included in a Cox regression analysis (CRA). Response to neoadjuvant therapy was characterized by a change in tumor thickness from pretherapy to preoperative scans using the modified RECIST criteria. Results In this study, 99 patients met the inclusion criteria. The median age of the included patients was 64.7 years, who were predominantly men, had smoking and asbestos exposure, and who received neoadjuvant therapy. The median change in tumor thickness following neoadjuvant therapy was -16.5% (interquartile range of -49.7% to +14.2%). CRA demonstrated reduced OS associated with non-epithelioid histology [hazard ratio (HR): 3.06, 95% confidence interval (CI): 1.62-5.78, p < 0.001] and a response to neoadjuvant therapy inferior to the median (HR: 2.70, CI: 1.55-4.72, p < 0.001). Patients who responded poorly (below median) to neoadjuvant therapy had lower median survival (15.8 months compared to 38.2 months, p < 0.001). Conclusion Poor response to neoadjuvant therapy in patients with MPM is associated with poor outcomes even following maximum surgical cytoreduction and should warrant a patient-centered discussion regarding goals of care and may therefore help guide further therapeutic decisions.
Collapse
Affiliation(s)
- Nathaniel Deboever
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nicolas Zhou
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Daniel J. McGrail
- Department of Bioinformatics and Computational Biology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Katarzyna Tomczak
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jacqueline L. Oliva
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hope A. Feldman
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Edwin Parra
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jianjun Zhang
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Percy P. Lee
- Department of Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mara B. Antonoff
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Wayne L. Hofstetter
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Reza J. Mehran
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ravi Rajaram
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - David C. Rice
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jack A. Roth
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Stephen S. Swisher
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ara A. Vaporciyan
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Mehmet Altan
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Annikka Weissferdt
- Department of Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Anne S. Tsao
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Cara L. Haymaker
- Department of Translational Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Boris Sepesi
- Department of Thoracic and Cardiovascular Surgery, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
17
|
Trono P, Tocci A, Palermo B, Di Carlo A, D'Ambrosio L, D'Andrea D, Di Modugno F, De Nicola F, Goeman F, Corleone G, Warren S, Paolini F, Panetta M, Sperduti I, Baldari S, Visca P, Carpano S, Cappuzzo F, Russo V, Tripodo C, Zucali P, Gregorc V, Marchesi F, Nistico P. hMENA isoforms regulate cancer intrinsic type I IFN signaling and extrinsic mechanisms of resistance to immune checkpoint blockade in NSCLC. J Immunother Cancer 2023; 11:e006913. [PMID: 37612043 PMCID: PMC10450042 DOI: 10.1136/jitc-2023-006913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Understanding how cancer signaling pathways promote an immunosuppressive program which sustains acquired or primary resistance to immune checkpoint blockade (ICB) is a crucial step in improving immunotherapy efficacy. Among the pathways that can affect ICB response is the interferon (IFN) pathway that may be both detrimental and beneficial. The immune sensor retinoic acid-inducible gene I (RIG-I) induces IFN activation and secretion and is activated by actin cytoskeleton disturbance. The actin cytoskeleton regulatory protein hMENA, along with its isoforms, is a key signaling hub in different solid tumors, and recently its role as a regulator of transcription of genes encoding immunomodulatory secretory proteins has been proposed. When hMENA is expressed in tumor cells with low levels of the epithelial specific hMENA11a isoform, identifies non-small cell lung cancer (NSCLC) patients with poor prognosis. Aim was to identify cancer intrinsic and extrinsic pathways regulated by hMENA11a downregulation as determinants of ICB response in NSCLC. Here, we present a potential novel mechanism of ICB resistance driven by hMENA11a downregulation. METHODS Effects of hMENA11a downregulation were tested by RNA-Seq, ATAC-Seq, flow cytometry and biochemical assays. ICB-treated patient tumor tissues were profiled by Nanostring IO 360 Panel enriched with hMENA custom probes. OAK and POPLAR datasets were used to validate our discovery cohort. RESULTS Transcriptomic and biochemical analyses demonstrated that the depletion of hMENA11a induces IFN pathway activation, the production of different inflammatory mediators including IFNβ via RIG-I, sustains the increase of tumor PD-L1 levels and activates a paracrine loop between tumor cells and a unique macrophage subset favoring an epithelial-mesenchymal transition (EMT). Notably, when we translated our results in a clinical setting of NSCLC ICB-treated patients, transcriptomic analysis revealed that low expression of hMENA11a, high expression of IFN target genes and high macrophage score identify patients resistant to ICB therapy. CONCLUSIONS Collectively, these data establish a new function for the actin cytoskeleton regulator hMENA11a in modulating cancer cell intrinsic type I IFN signaling and extrinsic mechanisms that promote protumoral macrophages and favor EMT. These data highlight the role of actin cytoskeleton disturbance in activating immune suppressive pathways that may be involved in resistance to ICB in NSCLC.
Collapse
Affiliation(s)
- Paola Trono
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Institute of Biochemistry and Cell Biology, Consiglio Nazionale delle Ricerche, Rome, Italy
| | - Annalisa Tocci
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Belinda Palermo
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Anna Di Carlo
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Lorenzo D'Ambrosio
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Daniel D'Andrea
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Francesca Di Modugno
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Frauke Goeman
- SAFU Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giacomo Corleone
- SAFU Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Sarah Warren
- NanoString Technologies Inc, Seattle, Washington, USA
| | - Francesca Paolini
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Mariangela Panetta
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Isabella Sperduti
- Biostatistics Unit, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Silvia Baldari
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Paolo Visca
- Pathology Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Silvia Carpano
- Second Division of Medical Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Federico Cappuzzo
- Second Division of Medical Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Vincenzo Russo
- Department of Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Claudio Tripodo
- Department of Health Sciences, Human Pathology Section, Tumor Immunology Unit, University of Palermo, Palermo, Italy
| | - Paolo Zucali
- Department of Oncology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Vanesa Gregorc
- Department of Oncology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Federica Marchesi
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Paola Nistico
- Tumor of Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
18
|
Müller B, Bärenwaldt A, Herzig P, Zippelius A, Maul LV, Hess V, König D, Läubli H. Changes of peripheral T cell subsets in melanoma patients with immune-related adverse events. Front Immunol 2023; 14:1125111. [PMID: 37122748 PMCID: PMC10130408 DOI: 10.3389/fimmu.2023.1125111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 03/31/2023] [Indexed: 05/02/2023] Open
Abstract
Introduction Immunotherapies have improved the prognosis of many cancer patients including patients with advanced melanoma. Immune checkpoint receptors including CTLA-4 and PD-1 have been established as main therapeutic targets for immunotherapy of melanoma. Although monotherapy is effective in melanoma patients, a dual therapy approach has been shown to be most effective. Dual checkpoint blockade, however, increases substantially the risk for immune-related adverse events (irAEs). Methods In this study, we characterized peripheral immune cell subsets in patients with anti-PD-1 monotherapy and with dual immune receptors blockade targeting PD-1 and CTLA-4. Results We found differences in peripheral T cells between patients who developed severe immune-related side effects and patients with mild irAEs. We identified several mainly changes in CD8+ T cell subsets in patients with severe irAE under dual PD-1 and CTLA-4 blockade. Discussion This work suggests that peripheral immune cell dynamics could be associated with severe immune-related side effects in patients receiving immune checkpoint inhibitors. These changes could be used as future biomarkers in early diagnosis of irAEs.
Collapse
Affiliation(s)
- Benjamin Müller
- Laboratory for Cancer Immunotherapy and Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Anne Bärenwaldt
- Laboratory for Cancer Immunotherapy and Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Petra Herzig
- Laboratory for Cancer Immunotherapy and Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Alfred Zippelius
- Laboratory for Cancer Immunotherapy and Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Oncology, University Hospital Basel, Basel, Switzerland
| | - Lara Valeska Maul
- Department of Dermatology, University Hospital Basel, Basel, Switzerland
| | - Viviane Hess
- Division of Oncology, University Hospital Basel, Basel, Switzerland
| | - David König
- Laboratory for Cancer Immunotherapy and Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Oncology, University Hospital Basel, Basel, Switzerland
| | - Heinz Läubli
- Laboratory for Cancer Immunotherapy and Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
- Division of Oncology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
19
|
Johnson ML, Strauss J, Patel MR, Garon EB, Eaton KD, Neskorik T, Morin J, Chao R, Halmos B. Mocetinostat in Combination With Durvalumab for Patients With Advanced NSCLC: Results From a Phase I/II Study. Clin Lung Cancer 2023; 24:218-227. [PMID: 36890020 DOI: 10.1016/j.cllc.2023.01.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/10/2023]
Abstract
BACKGROUND Histone deacetylase (HDAC) inhibitors have potential to augment the effectiveness of immune checkpoint inhibitors and overcome treatment resistance. This dose-escalation/expansion study (NCT02805660) investigated mocetinostat (class I/IV HDAC inhibitor) plus durvalumab in patients with advanced non-small cell lung cancer (NSCLC) across cohorts defined by tumor programmed death-ligand 1 (PD-L1) expression and prior experience with anti-programmed cell death protein-1 (anti-PD-1) or anti-PD-L1 regimens. PATIENTS AND METHODS Sequential cohorts of patients with solid tumors received mocetinostat (starting dose: 50 mg TIW) plus durvalumab at a standard dose (1500 mg Q4W) to determine the recommended phase II dose (RP2D: phase I primary endpoint), based on the observed safety profile. RP2D was administered to patients with advanced NSCLC across 4 cohorts grouped by tumor PD-L1 expression (none or low/high) and prior experience with anti-PD-L1 /anti-PD-1 agents (naïve, clinical benefit: yes/no). The phase II primary endpoint was objective response rate (ORR, RECIST v1.1). RESULTS Eighty-three patients were enrolled (phase I [n = 20], phase II [n = 63]). RP2D was mocetinostat 70 mg TIW plus durvalumab. ORR was 11.5% across the phase II cohorts, and responses were durable (median 329 days). Clinical activity was observed in NSCLC patients with disease refractory to prior checkpoint inhibitor treatment: ORR 23.1%. Across all patients, fatigue (41%), nausea (40%), and diarrhea (31%) were the most frequent treatment-related adverse events. CONCLUSION Mocetinostat 70 mg TIW plus durvalumab at the standard dose was generally well tolerated. Clinical activity was observed in patients with NSCLC unresponsive to prior anti-PD-(L)1 therapy.
Collapse
Affiliation(s)
| | | | - Manish R Patel
- Department of Medicine, Division of Hematology, Oncology, and Transplantation, University of Minnesota Masonic Cancer Center, Minneapolis, MN
| | - Edward B Garon
- Department of Medicine, David Geffen School of Medicine at UCLA, Santa Monica, CA
| | - Keith D Eaton
- Department of Medicine, Division of Medical Oncology, University of Washington and Fred Hutchinson Cancer Center, Seattle WA
| | - Tavette Neskorik
- Innovative Medicines Development, Mirati Therapeutics Inc., San Diego, CA
| | - Josée Morin
- Innovative Medicines Development, Mirati Therapeutics Inc., San Diego, CA
| | - Richard Chao
- Innovative Medicines Development, Mirati Therapeutics Inc., San Diego, CA
| | - Balazs Halmos
- Department of Oncology, Montefiore Medical Center, Bronx, NY.
| |
Collapse
|
20
|
Jing Y, Zeng H, Cheng R, Tian P, Li Y. [Advances of Immunotherapy Resistance and Coping Strategies
in Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2023; 26:66-77. [PMID: 36792083 PMCID: PMC9987066 DOI: 10.3779/j.issn.1009-3419.2023.102.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
Immunotherapy has significantly improved clinical outcomes of non-small cell lung cancer (NSCLC), however, along with the popularization of immunotherapy, immune resistance has become an unavoidable problem. Immunotherapy can induce extensive cellular and molecular alterations in the tumor microenvironment. Considering the mechanisms of immune resistance are not yet fully understood and the efficacy of standard chemotherapy regimens is limited, more effective coping strategies based on resistance mechanisms are urgently needed. In this review, we intend to summarize the known mechanisms of immune resistance and feasible strategies, so as to provide a foundation for clinicians to develop more individualized and precise regimens and finally improve patients' prognosis.
.
Collapse
Affiliation(s)
- Yawan Jing
- Department of Respiratory and Critical Care Medicine, West China Hospital, Chengdu 610041, China.,West China School of Medicine, Sichuan University, Chengdu 610041, China
| | - Hao Zeng
- Department of Respiratory and Critical Care Medicine, West China Hospital, Chengdu 610041, China.,West China School of Medicine, Sichuan University, Chengdu 610041, China
| | - Ruixin Cheng
- Department of Respiratory and Critical Care Medicine, West China Hospital, Chengdu 610041, China.,West China School of Medicine, Sichuan University, Chengdu 610041, China
| | - Panwen Tian
- Department of Respiratory and Critical Care Medicine, West China Hospital, Chengdu 610041, China.,Lung Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yalun Li
- Department of Respiratory and Critical Care Medicine, West China Hospital, Chengdu 610041, China.,Lung Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
21
|
Fintelmann FJ, Martin NA, Tahir I, Quinn EM, Allen TC, Joseph L, Nikolic B, Lee C. Optimizing molecular testing of lung cancer needle biopsy specimens: potential solutions from an interdisciplinary qualitative study. Respir Res 2023; 24:17. [PMID: 36650544 PMCID: PMC9847026 DOI: 10.1186/s12931-023-02321-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Molecular testing can detect actionable genomic alterations and tumor cell surface proteins in patients with non-small cell lung cancer (NSCLC). However, utilization remains suboptimal, representing missed treatment opportunities. This study aimed to identify challenges and potential solutions to obtaining percutaneous lung needle biopsy specimens for successful molecular testing in patients with advanced NSCLC. METHODS This interdisciplinary qualitative study included ten radiologists and four pathologists from academic and community settings across the United States who routinely perform and analyze percutaneous lung needle biopsies. Participants underwent semi-structured one-on-one interviews (Phase 1). Interview questionnaires were constructed based on a literature review of key lines of inquiry and conducted by professional market researchers using the theoretical domains framework. Primary barriers to molecular testing were identified using thematic analysis. Subsequently, multidisciplinary focus groups were convened to identify potential solutions (Phase 2). RESULTS Four themes emerged as barriers to molecular testing and were matched to the clinical workflow: (1) biopsy request, (2) biopsy procedure, (3) specimen analysis, and (4) communication. The nineteen potential solutions included adding a "checkbox" to indicate molecular testing in the biopsy request, leveraging pre-procedural imaging to guide biopsies, conserving tissue through appropriate allocation strategies and next generation sequencing panels instead of sequential single-gene assays, instituting reflex-molecular testing upon NSCLC diagnosis, tracking and communicating biopsy outcomes at multidisciplinary tumor boards, and improving integration of radiologists and pathologists into oncology care teams. CONCLUSIONS Potential solutions exist to increase successful molecular testing of lung needle biopsy specimens in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Florian J. Fintelmann
- grid.32224.350000 0004 0386 9924Department of Radiology, Division of Thoracic Imaging and Intervention, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114 USA
| | - Nikki A. Martin
- grid.443873.f0000 0004 0422 4933LUNGevity Foundation, Bethesda, MD USA
| | - Ismail Tahir
- grid.32224.350000 0004 0386 9924Department of Radiology, Division of Thoracic Imaging and Intervention, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114 USA
| | - Elissa M. Quinn
- grid.497611.c0000 0004 1794 1958Blueprint Medicines, Boston, MA USA
| | | | - Lija Joseph
- grid.461527.30000 0004 0383 4123Lowell General Hospital, Lowell, MA USA
| | - Boris Nikolic
- grid.439147.c0000 0004 0628 7583Wyoming Valley Radiology Associates, Wilkes-Barre General Hospital, Wilkes-Barre, PA USA
| | - Christopher Lee
- grid.50956.3f0000 0001 2152 9905Department of Imaging, Cedars-Sinai Medical Center, Los Angeles, CA USA
| |
Collapse
|
22
|
Grenda A, Iwan E, Krawczyk P, Frąk M, Chmielewska I, Bomba A, Giza A, Rolska-Kopińska A, Szczyrek M, Kieszko R, Kucharczyk T, Jarosz B, Wasyl D, Milanowski J. Attempting to Identify Bacterial Allies in Immunotherapy of NSCLC Patients. Cancers (Basel) 2022; 14:cancers14246250. [PMID: 36551735 PMCID: PMC9777223 DOI: 10.3390/cancers14246250] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Introduction: Factors other than PD-L1 (Programmed Death Ligand 1) are being sought as predictors for cancer immuno- or chemoimmunotherapy in ongoing studies and long-term observations. Despite high PD-L1 expression on tumor cells, some patients do not benefit from immunotherapy, while others, without the expression of this molecule, respond to immunotherapy. Attention has been paid to the composition of the gut microbiome as a potential predictive factor for immunotherapy effectiveness. Materials and Methods: Our study enrolled 47 Caucasian patients with stage IIIB or IV non-small cell lung cancer (NSCLC). They were eligible for treatment with first- or second-line immunotherapy or chemoimmunotherapy. We collected stool samples before the administration of immunotherapy. We performed next-generation sequencing (NGS) on DNA isolated from the stool sample and analyzed bacterial V3 and V4 of the 16S rRNA gene. Results: We found that bacteria from the families Barnesiellaceae, Ruminococcaceae, Tannerellaceae, and Clostridiaceae could modulate immunotherapy effectiveness. A high abundance of Bacteroidaaceae, Barnesiellaceae, and Tannerellaceae could extend progression-free survival (PFS). Moreover, the risk of death was significantly higher in patients with a high content of Ruminococcaceae family (HR = 6.3, 95% CI: 2.6 to 15.3, p < 0.0001) and in patients with a low abundance of Clostridia UCG-014 (HR = 3.8, 95% CI: 1.5 to 9.8, p = 0.005) regardless of the immunotherapy line. Conclusions: The Clostridia class in gut microbiota could affect the effectiveness of immunotherapy, as well as the length of survival of NSCLC patients who received this method of treatment.
Collapse
Affiliation(s)
- Anna Grenda
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8, 20-950 Lublin, Poland
- Correspondence: ; Tel.: +48-81-724-4293
| | - Ewelina Iwan
- Department of Omics Analyses, National Veterinary Research Institute, Partyzantow 57, 24-100 Pulawy, Poland
| | - Paweł Krawczyk
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8, 20-950 Lublin, Poland
| | - Małgorzata Frąk
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8, 20-950 Lublin, Poland
| | - Izabela Chmielewska
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8, 20-950 Lublin, Poland
| | - Arkadiusz Bomba
- Department of Omics Analyses, National Veterinary Research Institute, Partyzantow 57, 24-100 Pulawy, Poland
| | - Aleksandra Giza
- Department of Omics Analyses, National Veterinary Research Institute, Partyzantow 57, 24-100 Pulawy, Poland
| | - Anna Rolska-Kopińska
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8, 20-950 Lublin, Poland
| | - Michał Szczyrek
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8, 20-950 Lublin, Poland
| | - Robert Kieszko
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8, 20-950 Lublin, Poland
| | - Tomasz Kucharczyk
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8, 20-950 Lublin, Poland
| | - Bożena Jarosz
- Department of Neurosurgery and Pediatric Neurosurgery, Medical University of Lublin, Jaczewskiego 8, 20-950 Lublin, Poland
| | - Dariusz Wasyl
- Department of Omics Analyses, National Veterinary Research Institute, Partyzantow 57, 24-100 Pulawy, Poland
| | - Janusz Milanowski
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Jaczewskiego 8, 20-950 Lublin, Poland
| |
Collapse
|
23
|
Borgmann M, Oetting A, Meyer F, Möckelmann N, Droste C, von Bargen CM, Möller-Koop C, Witt M, Borgmann K, Rothkamm K, Betz C, Münscher A, Clauditz TS, Rieckmann T. The prognostic impact of B7-H3 and B7-H4 in head and neck squamous cell carcinoma. J Cancer Res Clin Oncol 2022:10.1007/s00432-022-04244-2. [PMID: 35941227 DOI: 10.1007/s00432-022-04244-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/29/2022] [Indexed: 10/15/2022]
Abstract
PURPOSE Immune checkpoint inhibition is a therapeutic option in many cancer entities. In head and neck squamous cell carcinoma (HNSCC) targeting of the PD-1/PD-L1 (B7-H1) axis is approved in recurrent/metastatic disease and is being explored in the curative setting. Here, we evaluated two related members of the B7 family, B7-H3 & B7-H4, for their prognostic impact under standard treatment. METHODS A tissue microarray (TMA) of a single center HNSCC cohort was stained for B7-H3 and B7-H4. Staining intensity and the number of tumor cells stained were assessed, and the expression was scored according to an established algorithm. Staining scores were correlated with clinicopathological parameters and associated with patient survival. mRNA levels of both proteins were associated with patient outcome using the TCGA dataset. RESULTS mRNA levels of B7-H3 and B7-H4 were not significantly associated with patient survival. TMA analysis revealed interpretable protein staining in 408 samples. Strong staining was the most frequent category for B7-H3 and no staining for B7-H4. In patients with p16-negative oropharyngeal SCC (OPSCC) and in a pooled cohort consisting of p16-negative OPSCC, laryngeal, hypopharyngeal and oral cavity SCC, strong B7-H3 expression was associated with better overall survival. For the latter cohort, this was in part due to reduced lymph node involvement. B7-H3 expression in p16-positive OPSCC and B7-H4 expression were not associated with outcome. CONCLUSION Despite a possible role in tumor immune escape, B7-H3 was associated with favorable prognosis in HPV-negative HNSCC in our cohort. The underlying mechanisms and a potential impact for B7-H3 targeting remain to be elucidated.
Collapse
Affiliation(s)
- Mara Borgmann
- Department of Otorhinolaryngology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Otorhinolaryngology, Asklepios Klinik Nord, Hamburg, Germany
| | - Agnes Oetting
- Department of Otorhinolaryngology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Felix Meyer
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nikolaus Möckelmann
- Department of Otorhinolaryngology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Otorhinolaryngology, Marienkrankenhaus Hamburg, Hamburg, Germany
| | - Conrad Droste
- University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Christina Möller-Koop
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Melanie Witt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kerstin Borgmann
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kai Rothkamm
- Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Betz
- Department of Otorhinolaryngology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Adrian Münscher
- Department of Otorhinolaryngology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Otorhinolaryngology, Marienkrankenhaus Hamburg, Hamburg, Germany
| | | | - Thorsten Rieckmann
- Department of Otorhinolaryngology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany. .,Department of Radiotherapy and Radiation Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|