1
|
Huang Z, Lou K, Qi M, Wang J, Li L, Sun B, Wang C, Zhou X, Chen D, Liu H. RERE-AS1 enhances the effect of CDK4/6 inhibitor Ribociclib and suppresses malignant phenotype in breast cancer via MEK/ERK pathway. J Transl Med 2024; 22:1052. [PMID: 39574120 PMCID: PMC11583401 DOI: 10.1186/s12967-024-05828-x] [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: 05/23/2024] [Accepted: 10/31/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Currently, there is a lack of biomarkers to identify breast cancer (BC) patients who would benefit from CDK4/6 inhibitors. This study combined machine learning (ML) algorithms based on transcriptomic data with both in vivo and in vitro experiments to identify therapeutic efficacy-related biomarkers of the CDK4/6 inhibitor ribociclib from the perspective of long non-coding RNA (lncRNA). METHODS We used the Genomics of Drug Sensitivity in Cancer database along with the "oncoPredict" algorithm to calculate the half maximal inhibitory concentration (IC50) values for ribociclib based on transcriptome data. ML algorithms were utilized to select key lncRNAs related to ribociclib and to establish a model which could be used for selection of potential beneficiaries of ribociclib. Cellular experiments were conducted to validate the ML analysis and explore the potential biological mechanisms by which RERE-AS1 influences ribociclib efficacy and malignant phenotype of BC cells. Correlation analysis with clinical pathological factors, RT-qPCR experiments on tissue specimens, and pan-cancer analysis were carried out to explore the expression pattern, and the prognostic and diagnostic potential of RERE-AS1 in cancers. RESULTS We have identified 11 key ribociclib-related lncRNAs and constructed an artificial neural network model (ANNM) based on lncRNA. Cellular experiments demonstrated that overexpression of RERE-AS1 promoted the anti-tumor activity of ribociclib in BC cells. Furthermore, RERE-AS1 is crucial in suppressing the malignant traits of BC cells through the reduction of MEK and ERK phosphorylation levels. Patients with smaller primary tumors and lower pathological stage exhibited higher levels of RERE-AS1 expression. Lastly, a pan-cancer analysis revealed that RERE-AS1 exhibits distinctly abnormal expression patterns, prognostic significance, and clinical diagnostic value in BC, compared to other cancers. CONCLUSIONS The ANNM established through ML algorithms can serve as predictive indicators for the efficacy of ribociclib in BC patients. LncRNA RERE-AS1, a newly discovered biomarker, holds significant promise for diagnosis, treatment, and enhancing the therapeutic response to ribociclib in BC.
Collapse
Affiliation(s)
- Zhidong Huang
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Kaixin Lou
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Mengyang Qi
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jinhui Wang
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Linwei Li
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Bo Sun
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Chen Wang
- The School of Basic Medicine, Tianjin Medical University, Tianjin, China
| | - Xirui Zhou
- The School of Basic Medicine, Tianjin Medical University, Tianjin, China
| | - Debo Chen
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China.
- Department of Breast Surgery, Quanzhou First Hospital of Fujian Medical University, Quanzhou, China.
| | - Hong Liu
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
2
|
TONG Y, LONG Y, ZHANG F, LI J. [Advances in Pseudoprogression of Immune Checkpoint Inhibitors
in Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2024; 27:306-320. [PMID: 38769834 PMCID: PMC11110244 DOI: 10.3779/j.issn.1009-3419.2024.101.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Indexed: 05/22/2024]
Abstract
The advent of immune checkpoint inhibitors (ICIs) has greatly improved the prognosis of advanced lung cancer patients, but can lead to pseudoprogression (PsP), which complicates clinical evaluation and management. PsP is manifested as temporary enlargement of the tumour or the appearance of new lesions, etc., and improvement in imaging occurs with continued treatment, mostly without worsening of clinical symptoms. Currently, there are still difficulties in the early diagnosis of PsP, and its occurrence mechanism is not yet clear, lacking good predictive factors and related biomarkers. This article reviews the current research status of PsP of ICIs in non-small cell lung cancer in order to provide helpful clinical strategies for oncologists using these drugs.
.
Collapse
|
3
|
Tsai YT, Schlom J, Donahue RN. Blood-based biomarkers in patients with non-small cell lung cancer treated with immune checkpoint blockade. J Exp Clin Cancer Res 2024; 43:82. [PMID: 38493133 PMCID: PMC10944611 DOI: 10.1186/s13046-024-02969-1] [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: 12/05/2023] [Accepted: 01/30/2024] [Indexed: 03/18/2024] Open
Abstract
The paradigm of non-small cell lung cancer (NSCLC) treatment has been profoundly influenced by the development of immune checkpoint inhibitors (ICI), but the range of clinical responses observed among patients poses significant challenges. To date, analyses of tumor biopsies are the only parameter used to guide prognosis to ICI therapy. Tumor biopsies, however, are often difficult to obtain and tissue-based biomarkers are limited by intratumoral heterogeneity and temporal variability. In response, there has been a growing emphasis on the development of "liquid biopsy"‒ derived biomarkers, which offer a minimally invasive means to dynamically monitor the immune status of NSCLC patients either before and/or during the course of treatment. Here we review studies in which multiple blood-based biomarkers encompassing circulating soluble analytes, immune cell subsets, circulating tumor DNA, blood-based tumor mutational burden, and circulating tumor cells have shown promising associations with the clinical response of NSCLC patients to ICI therapy. These investigations have unveiled compelling correlations between the peripheral immune status of patients both before and during ICI therapy and patient outcomes, which include response rates, progression-free survival, and overall survival. There is need for rigorous validation and standardization of these blood-based assays for broader clinical application. Integration of multiple blood-based biomarkers into comprehensive panels or algorithms also has the potential to enhance predictive accuracy. Further research aimed at longitudinal monitoring of circulating biomarkers is also crucial to comprehend immune dynamics and resistance mechanisms and should be used alongside tissue-based methods that interrogate the tumor microenvironment to guide treatment decisions and may inform on the development of novel therapeutic strategies. The data reviewed here reinforce the opportunity to refine patient stratification, optimize treatments, and improve outcomes not only in NSCLC but also in the wider spectrum of solid tumors undergoing immunotherapy.
Collapse
Affiliation(s)
- Yo-Ting Tsai
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey Schlom
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - Renee N Donahue
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
4
|
Masson C, Thouvenin J, Boudier P, Maillet D, Kuchler-Bopp S, Barthélémy P, Massfelder T. Biological Biomarkers of Response and Resistance to Immune Checkpoint Inhibitors in Renal Cell Carcinoma. Cancers (Basel) 2023; 15:3159. [PMID: 37370768 DOI: 10.3390/cancers15123159] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/09/2023] [Accepted: 06/10/2023] [Indexed: 06/29/2023] Open
Abstract
Renal cell carcinoma (RCC) represents around 2% of cancer-related deaths worldwide per year. RCC is an immunogenic malignancy, and treatment of metastatic RCC (mRCC) has greatly improved since the advent of the new immunotherapy agents, including immune checkpoint inhibitors (ICIs). However, it should be stressed that a large proportion of patients does not respond to these therapies. There is thus an urgent need to identify predictive biomarkers of efficacy or resistance associated with ICIs or ICI/Tyrosine kinase inhibitor (TKI) combinations; this is a major challenge to achieve precision medicine for mRCC in routine practice. To identify potential biomarkers, it is necessary to improve our knowledge on the biology of immune checkpoints. A lot of efforts have been made over the last decade in the field of immuno-oncology. We summarize here the main data obtained in this field when considering mRCC. As for clinical biomarkers, clinician and scientific experts of the domain are facing difficulties in identifying such molecular entities, probably due to the complexity of immuno-oncology and the constant adaptation of tumor cells to their changing environment.
Collapse
Affiliation(s)
- Claire Masson
- Regenerative NanoMedicine, Centre de Recherche en Biomédecine de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), UMR_S U1260 INSERM and University of Strasbourg, 67085 Strasbourg, France
| | - Jonathan Thouvenin
- Medical Oncology Department, Hospices Civils de Lyon, Hôpital Lyon Sud, 69310 Pierre-Bénite, France
| | - Philippe Boudier
- Medical Oncology Department, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France
| | - Denis Maillet
- Medical Oncology Department, Hospices Civils de Lyon, Hôpital Lyon Sud, 69310 Pierre-Bénite, France
| | - Sabine Kuchler-Bopp
- Regenerative NanoMedicine, Centre de Recherche en Biomédecine de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), UMR_S U1260 INSERM and University of Strasbourg, 67085 Strasbourg, France
| | - Philippe Barthélémy
- Medical Oncology Department, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France
| | - Thierry Massfelder
- Regenerative NanoMedicine, Centre de Recherche en Biomédecine de Strasbourg, Fédération de Médecine Translationnelle de Strasbourg (FMTS), UMR_S U1260 INSERM and University of Strasbourg, 67085 Strasbourg, France
| |
Collapse
|
5
|
Piro G, Carbone C, Agostini A, Esposito A, De Pizzol M, Novelli R, Allegretti M, Aramini A, Caggiano A, Granitto A, De Sanctis F, Ugel S, Corbo V, Martini M, Lawlor RT, Scarpa A, Tortora G. CXCR1/2 dual-inhibitor ladarixin reduces tumour burden and promotes immunotherapy response in pancreatic cancer. Br J Cancer 2023; 128:331-341. [PMID: 36385556 PMCID: PMC9902528 DOI: 10.1038/s41416-022-02028-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy with few therapeutic options available. Despite immunotherapy has revolutionised cancer treatment, the results obtained in PDAC are still disappointing. Emerging evidence suggests that chemokines/CXCRs-axis plays a pivotal role in immune tumour microenvironment modulation, which may influence immunotherapy responsiveness. Here, we evaluated the effectiveness of CXCR1/2 inhibitor ladarixin, alone or in combination with anti-PD-1, against immunosuppression in PDAC. METHODS A set of preclinical models was obtained by engrafting mouse PDAC-derived cells into syngeneic immune-competent mice, as well as by orthotopically transplanting patient-derived PDAC tumour into human immune-system-reconstituted (HIR) mice (HuCD34-NSG-mice). Tumour-bearing mice were randomly assigned to receive vehicles, ladarixin, anti-PD-1 or drugs combination. RESULTS CXCR1/2 inhibition by ladarixin reverted in vitro tumour-mediated M2 macrophages polarisation and migration. Ladarixin as single agent reduced tumour burden in cancer-derived graft (CDG) models with high-immunogenic potential and increased the efficacy of ICI in non-immunogenic CDG-resistant models. In a HIR mouse model bearing the immunogenic subtype of human PDAC, ladarixin showed high efficacy increasing the antitumor effect of anti-PD-1. CONCLUSION Ladarixin in combination with anti-PD-1 might represent an extremely effective approach for the treatment of immunotherapy refractory PDAC, allowing pro-tumoral to immune-permissive microenvironment conversion.
Collapse
Affiliation(s)
- Geny Piro
- Medical Oncology, Department of Medical and Surgical Sciences Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Carmine Carbone
- Medical Oncology, Department of Medical and Surgical Sciences Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Antonio Agostini
- Medical Oncology, Department of Medical and Surgical Sciences Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Annachiara Esposito
- Medical Oncology, Department of Medical and Surgical Sciences Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | | | - Rubina Novelli
- Dompé Farmaceutici S.p.A., Via Santa Lucia 6, Milan, Italy
| | | | - Andrea Aramini
- Dompé Farmaceutici S.p.A., Via Santa Lucia 6, Milan, Italy
| | - Alessia Caggiano
- Medical Oncology, Department of Medical and Surgical Sciences Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Alessia Granitto
- Division of Anatomic Pathology and Histology, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Francesco De Sanctis
- Department of Medicine, Section of Immunology, University of Verona, Verona, Italy
| | - Stefano Ugel
- Department of Medicine, Section of Immunology, University of Verona, Verona, Italy
| | - Vincenzo Corbo
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
- ARC-Net Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Maurizio Martini
- Division of Anatomic Pathology and Histology, Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy
| | - Rita Teresa Lawlor
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
- ARC-Net Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
- ARC-Net Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Giampaolo Tortora
- Medical Oncology, Department of Medical and Surgical Sciences Fondazione Policlinico Universitario "Agostino Gemelli" IRCCS, Rome, Italy.
- Medical Oncology, Department of Translational Medicine, Catholic University of the Sacred Heart, Rome, Italy.
| |
Collapse
|
6
|
Botticelli A, Pomati G, Cirillo A, Scagnoli S, Pisegna S, Chiavassa A, Rossi E, Schinzari G, Tortora G, Di Pietro FR, Cerbelli B, Di Filippo A, Amirhassankhani S, Scala A, Zizzari IG, Cortesi E, Tomao S, Nuti M, Mezi S, Marchetti P. The role of immune profile in predicting outcomes in cancer patients treated with immunotherapy. Front Immunol 2022; 13:974087. [PMID: 36405727 PMCID: PMC9671166 DOI: 10.3389/fimmu.2022.974087] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 09/22/2022] [Indexed: 11/06/2022] Open
Abstract
Background Despite the efficacy of immunotherapy, only a small percentage of patients achieves a long-term benefit in terms of overall survival. The aim of this study was to define an immune profile predicting the response to immune checkpoint inhibitors (ICIs). Methods Patients with advanced solid tumors, who underwent ICI treatment were enrolled in this prospective study. Blood samples were collected at the baseline. Thirteen soluble immune checkpoints, 3 soluble adhesion molecules, 5 chemokines and 11 cytokines were analyzed. The results were associated with oncological outcomes. Results Regardless of tumor type, patients with values of sTIM3, IFNα, IFNγ, IL1β, IL1α, IL12p70, MIP1β, IL13, sCD28, sGITR, sPDL1, IL10 and TNFα below the median had longer overall survival (p<0.05). By using cluster analysis and grouping the patients according to the trend of the molecules, two clusters were found. Cluster A had a significantly higher mean progression free survival (Cluster A=11.9 months vs Cluster B=3.5 months, p<0.01), a higher percentage of disease stability (Cluster A=34.5% vs. Cluster B=0%, p<0.05) and a lower percentage of disease progression (Cluster A=55.2% vs. Cluster B = 94.4%, p=0.04). Conclusion The combined evaluation of soluble molecules, rather than a single circulating factor, may be more suitable to represent the fitness of the immune system status in each patient and could allow to identify two different prognostic and predictive outcome profiles.
Collapse
Affiliation(s)
- Andrea Botticelli
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
| | - Giulia Pomati
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessio Cirillo
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
- *Correspondence: Alessio Cirillo,
| | - Simone Scagnoli
- Department of Medical and Surgical Sciences and Translational Medicine, University of Rome “Sapienza”, Rome, Italy
| | - Simona Pisegna
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
| | - Antonella Chiavassa
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
| | - Ernesto Rossi
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli Istituti di Ricovero e Cura a Carattere Scientifico (IRCSS), Rome, Italy
| | - Giovanni Schinzari
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli Istituti di Ricovero e Cura a Carattere Scientifico (IRCSS), Rome, Italy
- Medical Oncology, Universitá Cattolica del Sacro Cuore, Rome, Italy
| | - Giampaolo Tortora
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli Istituti di Ricovero e Cura a Carattere Scientifico (IRCSS), Rome, Italy
- Medical Oncology, Universitá Cattolica del Sacro Cuore, Rome, Italy
| | | | - Bruna Cerbelli
- Department of Medico-Surgical Sciences and Biotechnology, Polo Pontino, Sapienza University, Rome, Italy
| | - Alessandra Di Filippo
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, University of Rome “Sapienza”, Rome, Italy
| | - Sasan Amirhassankhani
- Department of Urology, S. Orsola-Malpighi Hospital University of Bologna, Via Palagi, Bologna, Italy
| | - Alessandro Scala
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli Istituti di Ricovero e Cura a Carattere Scientifico (IRCSS), Rome, Italy
| | - Ilaria Grazia Zizzari
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, University of Rome “Sapienza”, Rome, Italy
| | - Enrico Cortesi
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
| | - Silverio Tomao
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
| | - Marianna Nuti
- Laboratory of Tumor Immunology and Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, University of Rome “Sapienza”, Rome, Italy
| | - Silvia Mezi
- Department of Radiological, Oncological and Pathological Science, Sapienza University of Rome, Rome, Italy
| | | |
Collapse
|
7
|
Schindler H, Lusky F, Daniello L, Elshiaty M, Gaissmaier L, Benesova K, Souto-Carneiro M, Angeles AK, Janke F, Eichhorn F, Kazdal D, Schneider M, Liersch S, Klemm S, Schnitzler P, Stenzinger A, Sültmann H, Thomas M, Christopoulos P. Serum cytokines predict efficacy and toxicity, but are not useful for disease monitoring in lung cancer treated with PD-(L)1 inhibitors. Front Oncol 2022; 12:1010660. [PMID: 36387148 PMCID: PMC9662790 DOI: 10.3389/fonc.2022.1010660] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 10/14/2022] [Indexed: 01/09/2023] Open
Abstract
Introduction PD-(L)1 inhibitors (IO) have improved the prognosis of non-small-cell lung cancer (NSCLC), but more reliable predictors of efficacy and immune-related adverse events (irAE) are urgently needed. Cytokines are important effector molecules of the immune system, whose potential clinical utility as biomarkers remains unclear. Methods Serum samples from patients with advanced NSCLC receiving IO either alone in the first (1L, n=46) and subsequent lines (n=50), or combined with chemotherapy (ICT, n=108) were analyzed along with age-matched healthy controls (n=15) at baseline, after 1 and 4 therapy cycles, and at disease progression (PD). Patients were stratified in rapid progressors (RP, progression-free survival [PFS] <120 days), and long-term responders (LR, PFS >200 days). Cytometric bead arrays were used for high-throughput quantification of 20 cytokines and other promising serum markers based on extensive search of the current literature. Results Untreated NSCLC patients had increased levels of various cytokines and chemokines, like IL-6, IL-8, IL-10, CCL5, G-CSF, ICAM-1, TNF-RI and VEGF (fold change [FC]=1.4-261, p=0.026-9x10-7) compared to age-matched controls, many of which fell under ICT (FC=0.2-0.6, p=0.014-0.002), but not under IO monotherapy. Lower baseline levels of TNF-RI were associated with longer PFS (hazard ratio [HR]= 0.42-0.54; p=0.014-0.009) and overall survival (HR=0.28-0.34, p=0.004-0.001) after both ICT and IO monotherapy. Development of irAE was associated with higher baseline levels of several cytokines, in particular of IL-1β and angiogenin (FC=7-9, p=0.009-0.0002). In contrast, changes under treatment were very subtle, there were no serum correlates of radiologic PD, and no association between dynamic changes in cytokine concentrations and clinical outcome. No relationship was noted between the patients' serologic CMV status and serum cytokine levels. Conclusions Untreated NSCLC is characterized by increased blood levels of several pro-inflammatory and angiogenic effectors, which decrease under ICT. Baseline serum cytokine levels could be exploited for improved prediction of subsequent IO benefit (in particular TNF-RI) and development of irAE (e.g. IL-1β or angiogenin), but they are not suitable for longitudinal disease monitoring. The potential utility of IL-1/IL-1β inhibitors in the management and/or prevention of irAE in NSCLC warrants investigation.
Collapse
Affiliation(s)
- Hannah Schindler
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, Heidelberg, Germany,Translational Lung Research Center Heidelberg (TLRC-H), member of the German Center of Lung Research (DZL), Heidelberg, Germany
| | - Fabienne Lusky
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, Heidelberg, Germany,Translational Lung Research Center Heidelberg (TLRC-H), member of the German Center of Lung Research (DZL), Heidelberg, Germany
| | - Lea Daniello
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, Heidelberg, Germany,Translational Lung Research Center Heidelberg (TLRC-H), member of the German Center of Lung Research (DZL), Heidelberg, Germany
| | - Mariam Elshiaty
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, Heidelberg, Germany,Translational Lung Research Center Heidelberg (TLRC-H), member of the German Center of Lung Research (DZL), Heidelberg, Germany
| | - Lena Gaissmaier
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, Heidelberg, Germany,Translational Lung Research Center Heidelberg (TLRC-H), member of the German Center of Lung Research (DZL), Heidelberg, Germany
| | - Karolina Benesova
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Margarida Souto-Carneiro
- Department of Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Arlou Kristina Angeles
- Division of Cancer Genome Research (B063), German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Florian Janke
- Division of Cancer Genome Research (B063), German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Florian Eichhorn
- Department of Thoracic Surgery, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Daniel Kazdal
- Translational Lung Research Center Heidelberg (TLRC-H), member of the German Center of Lung Research (DZL), Heidelberg, Germany,Department of Molecular Pathology Institute of Pathology Heidelberg, Heidelberg, Germany
| | - Marc Schneider
- Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Stephan Liersch
- Department of Pharmacy, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, Heidelberg, Germany
| | - Sarah Klemm
- Center for Infectious Diseases, Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Paul Schnitzler
- Center for Infectious Diseases, Virology, University Hospital Heidelberg, Heidelberg, Germany
| | - Albrecht Stenzinger
- Translational Lung Research Center Heidelberg (TLRC-H), member of the German Center of Lung Research (DZL), Heidelberg, Germany,Department of Molecular Pathology Institute of Pathology Heidelberg, Heidelberg, Germany
| | - Holger Sültmann
- Translational Lung Research Center Heidelberg (TLRC-H), member of the German Center of Lung Research (DZL), Heidelberg, Germany,Division of Cancer Genome Research (B063), German Cancer Research Center (DKFZ) and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Michael Thomas
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, Heidelberg, Germany,Translational Lung Research Center Heidelberg (TLRC-H), member of the German Center of Lung Research (DZL), Heidelberg, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik and National Center for Tumor Diseases (NCT) at Heidelberg University Hospital, Heidelberg, Germany,Translational Lung Research Center Heidelberg (TLRC-H), member of the German Center of Lung Research (DZL), Heidelberg, Germany,*Correspondence: Petros Christopoulos,
| |
Collapse
|
8
|
Ghita I, Piperi E, Atamas SP, Bentzen SM, Ord RA, Dyalram D, Lubek JE, Younis RH. Cytokine profiling in plasma distinguishes the histological inflammatory subtype of head and neck squamous cell carcinoma and a novel regulatory role of osteopontin. FRONTIERS IN ORAL HEALTH 2022; 3:993638. [PMID: 36338570 PMCID: PMC9632968 DOI: 10.3389/froh.2022.993638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 08/18/2022] [Indexed: 12/05/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) can be classified according to the histological inflammatory subtype (HIS) into inflamed (HIS-INF) or immune excluded (HIS-IE). HIS-IE was previously associated with higher levels of soluble Semaphorin 4D (HsS4D) in plasma, and higher transcriptional levels of osteopontin (OPN) in the tumor tissue, compared to HIS-INF. The goal of the current study is to investigate whether the HIS inflammatory subtype can be distinguished by a differential cytokine panel in peripheral blood. Retrospectively collected five HIS-INF and five HIS-IE tumor tissue with paired plasma were included in the study. Five healthy donors (HD) and five autoimmune/chronic inflammatory conditions (AI/CI) were controls. The ELISA-Luminex™ system was used to detect 40 traditional cytokines in plasma. Human cytokine array (104 cytokines) was used for the conditioned medium (CM) of the HNSCC HN6 cell line. Semaphorin 4D (Sema4D) siRNA and recombinant human osteopontin (rh-OPN) were used to investigate the effect of OPN on Sema4D expression. The HIS-IE cytokine profile was higher than HIS-INF but comparable to AI/CI. HIS-INF had the lowest cytokine levels. HIS-IE was differentially higher in IP-10 and IL8 compared to HD, while HIS-INF was higher in IL-10. Sema4D inhibition in HN6 resulted in a decrease of OPN in the CM of HN6, and treatment with rh-OPN rescued Sema4D in HN6 cell lysate and associated CM. In conclusion, the current work demonstrates a novel association between the HIS subtypes and a differential pattern of cytokine expression in plasma. These findings can open new avenues for HNSCC patient stratification and hence provide better personalized treatment.
Collapse
Affiliation(s)
- Ioana Ghita
- Department of Oncology and Diagnostic Sciences, Division of Oral and Maxillofacial Pathology, University of Maryland School of Dentistry, Baltimore, MD, United States
| | - Evangelia Piperi
- Department of Oncology and Diagnostic Sciences, Division of Oral and Maxillofacial Pathology, University of Maryland School of Dentistry, Baltimore, MD, United States
- Department of Oral Medicine / Pathology and Hospital Dentistry, School of Dentistry, National and Kapodistrian University of Athens, Athens, Greece
| | - Sergei P. Atamas
- Department of Medicine, Division of Rheumatology and Clinical Immunology, University of Maryland School of Medicine. Baltimore, MD, United States
| | - Soren M. Bentzen
- Department of Epidemiology and Public Health, Division of Biostatistics and Bioinformatics, University of Maryland School of Medicine. Baltimore, MD, United States
- Biostatistics Core, Institute of Clinical and Translational Research, University of Maryland, Baltimore, MD, United States
- Biostatistics Division, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Robert A. Ord
- Department of Oral and Maxillofacial Surgery, University of Maryland School of Dentistry, Baltimore, MD, United States
- Head and Neck Surgery Department of Oral and Maxillofacial Surgery, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Donita Dyalram
- Department of Oral and Maxillofacial Surgery, University of Maryland School of Dentistry, Baltimore, MD, United States
- Head and Neck Surgery Department of Oral and Maxillofacial Surgery, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Joshua E. Lubek
- Department of Oral and Maxillofacial Surgery, University of Maryland School of Dentistry, Baltimore, MD, United States
- Head and Neck Surgery Department of Oral and Maxillofacial Surgery, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| | - Rania H. Younis
- Department of Oncology and Diagnostic Sciences, Division of Oral and Maxillofacial Pathology, University of Maryland School of Dentistry, Baltimore, MD, United States
- Division of Tumor immunology and Immunotherapy, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, United States
| |
Collapse
|
9
|
Rizzo M, Varnier L, Pezzicoli G, Pirovano M, Cosmai L, Porta C. IL-8 and its role as a potential biomarker of resistance to anti-angiogenic agents and immune checkpoint inhibitors in metastatic renal cell carcinoma. Front Oncol 2022; 12:990568. [PMID: 36059687 PMCID: PMC9437355 DOI: 10.3389/fonc.2022.990568] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Accepted: 08/01/2022] [Indexed: 11/24/2022] Open
Abstract
The therapeutic armamentarium of metastatic Renal Cell Carcinoma (mRCC) has consistently expanded in recent years, with the introduction of VEGF/VEGFR (Vascular Endothelial Growth Factor/Vascular Endothelial Growth Factor Receptor) inhibitors, mTOR (mammalian Target Of Rapamycin) inhibitors and Immune Checkpoint (IC) inhibitors. Currently, for the first-tline treatment of mRCC it is possible to choose between a VEGFR-TKI (VEGFR-Tyrosine Kinase Inhibitor) monotherapy, an ICI-ICI (Immune Checkpoint Inhibitor) combination and an ICI-VEGFRTKI combination. However, a consistent part of patients does not derive benefit from first-line therapy with ICIs; moreover, the use of combination regimens exposes patients to significant toxicities. Therefore, there is a critical need to develop prognostic and predictive biomarkers of response to VEGFR-TKIs and ICIs, and measurement of serum IL-8 is emerging as a potential candidate in this field. Recent retrospective analyses of large phase II and phase III trials found that elevated baseline serum IL-8 correlated with higher levels of tumor and circulating immunosuppressive myeloid cells, decreased T cell activation and poor response to treatment. These findings must be confirmed in prospective clinical trials; however, they provide evidence for a potential use of serum IL-8 as biomarker of resistance to VEGFR-TKIs and ICIs. Considering the amount of new agents and treatment regimens which are transforming the management of metastatic renal cell carcinoma, serum IL-8 could become a precious resource in tailoring the best therapy for each individual patient with the disease.
Collapse
Affiliation(s)
- Mimma Rizzo
- Division of Medical Oncology, Azienda Ospedaliero Universitaria Consorziale Policlinico di Bari, Bari, Italy
- *Correspondence: Mimma Rizzo,
| | - Luca Varnier
- Department of Pediatrics, Meyer’ Childrens University Hospital, Florence, Italy
| | - Gaetano Pezzicoli
- Department of Interdisciplinary Medicine, School of Medicine, University of Bari “A. Moro”, Bari, Italy
| | - Marta Pirovano
- Division of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale (ASST) Fatebenefratelli-Sacco, Fatebenefratelli Hospital, Milan, Italy
| | - Laura Cosmai
- Division of Nephrology and Dialysis, Azienda Socio-Sanitaria Territoriale (ASST) Fatebenefratelli-Sacco, Fatebenefratelli Hospital, Milan, Italy
| | - Camillo Porta
- Division of Medical Oncology, Azienda Ospedaliero Universitaria Consorziale Policlinico di Bari, Bari, Italy
- Chair of Oncology, Interdisciplinary Department of Medicine, University of Bari “A. Moro”, Bari, Italy
| |
Collapse
|