1
|
Rudzińska A, Juchaniuk P, Oberda J, Krukowska K, Krzyśkowska S, Kuchta E, Rodzajewska A, Janiszewska M, Machulska-Ciuraj K, Szklener K. Moderate toxicity with late onset as a good omen: association between toxicity and survival in the checkpoint inhibitor immunotherapy-a single center experience. Front Immunol 2025; 16:1527103. [PMID: 40356892 PMCID: PMC12066656 DOI: 10.3389/fimmu.2025.1527103] [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: 11/12/2024] [Accepted: 03/17/2025] [Indexed: 05/15/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy by enhancing T-cell-mediated immune responses against tumors. However, their use can lead to immune-related adverse events (irAEs) impacting patient outcomes. This single-center, observational study investigates the relationship between immune-related adverse events (irAEs) and survival outcomes and, to our knowledge, is the first of this kind in Polish population. Data of the 151 patients treated with ICIs, with or without chemotherapy, at the Department of Clinical Oncology and Chemotherapy in the Independent Public Hospital No. 4 in Lublin were collected from electronic medical records. Statistical analyses were performed using the Kaplan-Meier estimator, log-rank test, and multivariable Cox proportional hazard model (p < 0.05). IrAEs were observed in 38% of the patients, with the most common being thyroid dysfunction (11.9%) and dermal toxicity (6.6%). The median OS for patients with irAEs was 18.7 months, compared to 13.6 months for those without irAEs, though the difference was not statistically significant (p = 0.284). Patients with moderate toxicity had the highest median OS (26 months), while those with severe toxicity had a median OS of 6.41 months. Late-onset irAEs were associated with improved OS and PFS. Pack-years of smoking significantly impacted both OS (HR = 1.01, p = 0.014) and PFS (HR = 1.01, p = 0.011). Despite results not reaching statistical significance, the findings emphasize the clinical relevance of irAEs in treatment optimization and warrant further research to better understand their role in patient outcomes.
Collapse
Affiliation(s)
- Anna Rudzińska
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, Lublin, Poland
| | - Pola Juchaniuk
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, Lublin, Poland
| | - Jakub Oberda
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, Lublin, Poland
| | - Kamila Krukowska
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, Lublin, Poland
| | - Sylwia Krzyśkowska
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, Lublin, Poland
| | - Eliza Kuchta
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, Lublin, Poland
| | - Anna Rodzajewska
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, Lublin, Poland
| | - Mariola Janiszewska
- Department of Medical Informatics and Statistics with e-Health Lab, Medical University of Lublin, Lublin, Poland
| | | | - Katarzyna Szklener
- Department of Clinical Oncology and Chemotherapy, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
2
|
Cao LM, Yu YF, Li ZZ, Wang GR, Xiao Y, Luo HY, Liu B, Bu LL. Neoadjuvant Chemoimmunotherapy for Resectable Head and Neck Squamous Cell Carcinoma: Systematic Review and Meta-analysis. Ann Surg Oncol 2025:10.1245/s10434-025-17195-y. [PMID: 40102288 DOI: 10.1245/s10434-025-17195-y] [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: 12/05/2024] [Accepted: 03/03/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Neoadjuvant chemoimmunotherapy provides a good pathological response in patients with resectable head and neck squamous cell carcinoma (HNSCC). Currently, there is no comprehensive systematic review that rigorously evaluates and summarizes the existing studies. In this study, we aimed to synthesize the results on the efficacy of neoadjuvant chemoimmunotherapy in resectable HNSCC to obtain higher-level evidence. METHODS The PubMed, Web of Science, Scopus, and Academic Search Complete (EBSCO) databases, along with ClinicalTrials.gov, Google Scholar, and conference abstracts, were comprehensively searched. The publication dates of the literature were limited to January 2015-July 2024. Meta-analysis was performed using a random-effects model. The percentage of major pathological response (MPR), pathological complete response (pCR), and overall disease-free survival (DFS) were synthesized. The odds ratios of a combined positive score (CPS) ≥ 20 for MPR and the diagnostic performance of using radiological objective response to determine MPR were further explored. RESULTS A total of 13 studies with 458 patients who received neoadjuvant chemoimmunotherapy and 443 patients who underwent curative surgery were included. The pooled MPR, pCR, and overall DFS rates were 61%, 37%, and 91%, respectively. The odds ratios of a CPS ≥ 20 for achieving MPR was 2.09 compared with those with a CPS < 20. The sensitivity of using radiological objective response to determine MPR was 0.91 and the specificity was 0.46, with an area under the curve of 0.76. CONCLUSION Neoadjuvant chemoimmunotherapy showed promising results for resectable HNSCC. A CPS ≥ 20 can be used to screen for treatment-sensitive patients, and radiological examinations can be used to detect pathological response. Definitive conclusions require data from longer follow-up periods and controlled studies.
Collapse
Affiliation(s)
- Lei-Ming Cao
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yi-Fu Yu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yao Xiao
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Han-Yue Luo
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Bing Liu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral and Maxillofacial Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral and Maxillofacial Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
3
|
Yu Z, Dong C, Yang Y, Zheng Z, Ge X. USP21 stabilizes immune checkpoint of CD276 and serves as an immunological and tumor prognostic biomarker. Biochem Biophys Res Commun 2025; 745:151221. [PMID: 39736236 DOI: 10.1016/j.bbrc.2024.151221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/01/2025]
Abstract
Ubiquitin-specific protease 21 (USP21) belongs to the ubiquitin-specific protease family and is a member of the deubiquitinating enzyme (DUB) family. Previous research has shown that USP21 promotes cancer initiation and progression. However, there have been few pan-cancer analysis on USP21. We analyzed the expression levels of USP21 mRNA and protein in various human tumor tissues using several public databases such as The Cancer Genome Atlas (TCGA), Genotype Tissue Expression (GTEx), and Human Protein Atlas (HPA). Kaplan-Meier survival analyses were utilized to test the effect of USP21 on overall survival (OS) and progression-free interval (PFS) of these tumor patients. Our study demonstrated that USP21 was differentially expressed between normal and malignant tissues, conferring a notable value in evaluation of prognosis and diagnosis. In addition, enrichment and correlation analyses linking USP21 with immune features such as immune-cell-infiltration rate and immune-checkpoint-gene expression indicated that USP21 is an applicable immunotherapeutic marker for liver cancer. To further elucidate the role of USP21, we downregulated its expression in hepatocellular carcinoma cells and identified a remarkable decrease in expression of the immune checkpoint CD276, which contributes to the immune escape of tumor cells by suppressing the immune system. Together, our results indicated a promising potential of USP21 for future tumor prevention.
Collapse
Affiliation(s)
- Zhu Yu
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Chengyuan Dong
- School of Medicine, Anhui University of Science and Technology, Huainan, China
| | - Yanrong Yang
- Tongji University Cancer Center, School of Medicine, Tongji University, Shanghai, China
| | - Zening Zheng
- Tongji University Cancer Center, School of Medicine, Tongji University, Shanghai, China
| | - Xin Ge
- Department of Clinical Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
4
|
Zhang J, Wei Q, Piao Y, Shao S, Zhou Z, Tang J, Xiang J, Shen Y. Synergistic Combination of Oral Transcytotic Nanomedicine and Histone Demethylase Inhibitor for Enhanced Cancer Chemoimmunotherapy. ACS NANO 2024; 18:33729-33742. [PMID: 39612220 DOI: 10.1021/acsnano.4c14816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
Oral nanomedicines present a preferable avenue for cancer immunotherapy, but their efficacy is limited by gastrointestinal absorption challenges, tumor physiopathologic barriers, and immune evasion mechanisms. Here, we present an approach that combines an oral transcytotic doxorubicin (DOX) nanomedicine with the histone demethylase inhibitor 5-carboxy-8-hydroxyquinoline (IOX1), thereby enabling synergistic chemoimmunotherapy. We demonstrate that IOX1 significantly augments the transcytosis capabilities of DOX-loaded poly(2-(N-oxide-N,N-diethylamino)ethylmethacrylate)-poly(ε-caprolactone) micelles (OPDOX), promoting their transcellular transport across various cellular barriers (villus, endothelial, and tumor cells), thus improving oral adsorption, vascular extravasation, and tumor penetration. Furthermore, IOX1 sensitizes chemotherapy to potentiate DOX-induced immunogenic cell death and downregulates programmed cell death-ligand 1 to disrupt the immune checkpoint mechanism, synergistically boosting robust antitumor immune responses. Consequently, orally administered OPDOX in combination with IOX1 efficiently inhibits CT26 tumor growth, highlighting the significant potential for enhancing the efficacy of oral nanomedicines in cancer chemoimmunotherapy.
Collapse
Affiliation(s)
- Jing Zhang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Qiuyu Wei
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Shiqun Shao
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Jiajia Xiang
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Center for Bionanoengineering, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
- Key Laboratory of Biomass Chemical Engineering of the Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
5
|
Chelushkin MA, van Dorp J, van Wilpe S, Seignette IM, Mellema JJJ, Alkemade M, Gil-Jimenez A, Peters D, Brugman W, Stockem CF, Hooijberg E, Broeks A, van Rhijn BWG, Mertens LS, van der Heijden AG, Mehra N, van Montfoort ML, Wessels LFA, Vis DJ, van der Heijden MS. Platinum-Based Chemotherapy Induces Opposing Effects on Immunotherapy Response-Related Spatial and Stromal Biomarkers in the Bladder Cancer Microenvironment. Clin Cancer Res 2024; 30:4227-4239. [PMID: 39047168 DOI: 10.1158/1078-0432.ccr-24-0724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/08/2024] [Accepted: 07/23/2024] [Indexed: 07/27/2024]
Abstract
PURPOSE Platinum-based chemotherapy and immune checkpoint inhibitors are key components of systemic treatment for muscle-invasive and advanced urothelial cancer. The ideal integration of these two treatment modalities remains unclear as clinical trials have led to inconsistent results. Modulation of the tumor-immune microenvironment by chemotherapy is poorly characterized. We aimed to investigate this modulation, focusing on potential clinical implications for immune checkpoint inhibitor response. EXPERIMENTAL DESIGN We assessed immune cell densities, spatial relations, and tumor/stromal components from 116 patients with urothelial bladder cancer (paired data for 95 patients) before and after platinum-based chemotherapy. RESULTS Several published biomarkers for immunotherapy response changed upon chemotherapy treatment. The intratumoral CD8+ T-cell percentage increased after treatment and was associated with increased TNFα-via-NF-κB signaling. The percentage of PDL1+ immune cells was higher after chemotherapy. An increase in chemo-induced changes that potentially inhibit an antitumor immune response was also observed, including increased fibroblast-based TGFβ signaling and distances from immune cells to the nearest cancer cell. The latter two parameters correlated significantly in posttreatment samples, suggesting that TGFβ signaling in fibroblasts may play a role in spatially separating immune cells from cancer cells. We examined specific chemotherapy regimens and found that treatment with methotrexate, vinblastine, doxorubicin, and cisplatin was associated with an increase in the macrophage cell percentage. Gemcitabine-containing chemotherapy was associated with upregulation of fibroblast TGFβ signaling. CONCLUSIONS The opposing effects of platinum-based chemotherapy on the immune cell composition and stromal context of the tumor-immune microenvironment may explain the inconsistent results of clinical trials investigating chemotherapy and immune checkpoint inhibitor combinations in bladder cancer.
Collapse
Affiliation(s)
- Maksim A Chelushkin
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Tumor Biology and Immunology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Jeroen van Dorp
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sandra van Wilpe
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Iris M Seignette
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jan-Jaap J Mellema
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Maartje Alkemade
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Alberto Gil-Jimenez
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Dennis Peters
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Wim Brugman
- Genomics Core Facility, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Chantal F Stockem
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Erik Hooijberg
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Annegien Broeks
- Core Facility Molecular Pathology & Biobanking, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Bas W G van Rhijn
- Department of Urology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Urology, Caritas St. Josef Medical Center, University of Regensburg, Regensburg, Germany
| | - Laura S Mertens
- Department of Urology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Niven Mehra
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, the Netherlands
| | | | - Lodewyk F A Wessels
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
- Faculty of Electrical Engineering, Mathematics and Computer Science, Delft University of Technology, Delft, the Netherlands
| | - Daniel J Vis
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - Michiel S van der Heijden
- Department of Molecular Carcinogenesis, The Netherlands Cancer Institute, Amsterdam, the Netherlands
- Department of Medical Oncology, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
6
|
Lim SY, Kim L, Kim H, Park JA, Yun J, Lim KS. Synergistic Chemo-Immunotherapy: Recombinant Fusion Protein-Based Surface Modification of NK Cell for Targeted Cancer Treatment. Pharmaceutics 2024; 16:1189. [PMID: 39339225 PMCID: PMC11435017 DOI: 10.3390/pharmaceutics16091189] [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: 07/25/2024] [Revised: 09/01/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
While traditional combination anticancer treatments have shown promising results, there remains significant interest in developing innovative methods to enhance and integrate chemotherapy and immunotherapy. This study introduces a recombinant fusion protein-based cell surface modification system that synergistically combines chemotherapy and immunotherapy into a single-targeted chemo-immunotherapy approach. A cell surface-modified protein composed of an antibody-specific binding domain and a cell-penetrating domain rapidly converts immune cells into chemo-immuno therapeutics by binding to antibodies on the surface of immune cells. Utilizing a non-invasive, non-toxic approach free of chemical modifications and binding, our system homogeneously transforms immune cells by transiently introducing targeted cytotoxic drugs into them. The surface-engineered immune cells loaded with antibody-drug conjugates (ADCs) significantly inhibit the growth of target tumors and enhance the targeted elimination of cancer cells. Therefore, NK cells modified by the cell surface-modified protein to incorporate ADCs could be expected to achieve the combined effects of targeted cancer cell recognition, chemotherapy, and immunotherapy, thereby enhancing their therapeutic efficacy against cancer. This strategy allows for the efficient and rapid preparation of advanced chemo-immuno therapeutics to treat various types of cancer and provides significant potential to improve the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Su Yeon Lim
- Department of Smart Health Science and Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Luna Kim
- Department of Smart Health Science and Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Hongbin Kim
- Department of Smart Health Science and Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Jeong-Ann Park
- Department of Environmental Engineering, College of Art, Culture and Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Jina Yun
- Division of Hemato-Oncology, Department of Medicine, Soonchunhyang University Bucheon Hospital, Bucheon 14584, Republic of Korea
| | - Kwang Suk Lim
- Department of Smart Health Science and Technology, Kangwon National University, Chuncheon 24341, Republic of Korea
- Department of Biotechnology and Bioengineering, College of Art, Culture and Engineering, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
7
|
Jin H, Wang Y, Li X, Yang Y, Qi R. Radiomics nomogram for predicting chemo-immunotherapy efficiency in advanced non-small cell lung cancer. Sci Rep 2024; 14:20788. [PMID: 39242619 PMCID: PMC11379930 DOI: 10.1038/s41598-024-63415-y] [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: 09/05/2023] [Accepted: 05/28/2024] [Indexed: 09/09/2024] Open
Abstract
This study aimed to explore potential radiomics biomarkers in predicting the efficiency of chemo-immunotherapy in patients with advanced non-small cell lung cancer (NSCLC). Eligible patients were prospectively assigned to receive chemo-immunotherapy, and were divided into a primary cohort (n = 138) and an internal validation cohort (n = 58). Additionally, a separative dataset was used as an external validation cohort (n = 60). Radiomics signatures were extracted and selected from the primary tumor sites from chest CT images. A multivariate logistic regression analysis was conducted to identify the independent clinical predictors. Subsequently, a radiomics nomogram model for predicting the efficiency of chemo-immunotherapy was conducted by integrating the selected radiomics signatures and the independent clinical predictors. The receiver operating characteristic (ROC) curves demonstrated that the radiomics model, the clinical model, and the radiomics nomogram model achieved areas under the curve (AUCs) of 0.85 (95% confidence interval [CI] 0.78-0.92), 0.76 (95% CI 0.68-0.84), and 0.89 (95% CI 0.84-0.94), respectively, in the primary cohort. In the internal validation cohort, the corresponding AUCs were 0.93 (95% CI 0.86-1.00), 0.79 (95% CI 0.68-0.91), and 0.96 (95% CI 0.90-1.00) respectively. Moreover, in the external validation cohort, the AUCs were 0.84 (95% CI 0.72-0.96), 0.75 (95% CI 0.62-0.87), and 0.86 (95% CI 0.75-0.96), respectively. In conclusion, the radiomics nomogram provides a convenient model for predicting the effect of chemo-immunotherapy in advanced NSCLC patients.
Collapse
Affiliation(s)
- Hua Jin
- Department of Respiratory Medicine, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Yuchao Wang
- Department of Medical Imaging, Third Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150030, China.
| | - Xushuo Li
- Department of Clinical Laboratory, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Ying Yang
- Department of Clinical Laboratory, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Ruixue Qi
- Department of Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai, 201508, China.
| |
Collapse
|
8
|
Rakhmilevich AL, Tsarovsky NW, Felder M, Zaborek J, Moram S, Erbe AK, Pieper AA, Spiegelman DV, Cheng EM, Witt CM, Overwijk WW, Morris ZS, Sondel PM. A combined radio-immunotherapy regimen eradicates late-stage tumors in mice. Front Immunol 2024; 15:1419773. [PMID: 39076988 PMCID: PMC11284032 DOI: 10.3389/fimmu.2024.1419773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 07/02/2024] [Indexed: 07/31/2024] Open
Abstract
Background The majority of experimental approaches for cancer immunotherapy are tested against relatively small tumors in tumor-bearing mice, because in most cases advanced cancers are resistant to the treatments. In this study, we asked if even late-stage mouse tumors can be eradicated by a rationally designed combined radio-immunotherapy (CRI) regimen. Methods CRI consisted of local radiotherapy, intratumoral IL-12, slow-release systemic IL-2 and anti- CTLA-4 antibody. Therapeutic effects of CRI against several weakly immunogenic and immunogenic mouse tumors including B78 melanoma, MC38 and CT26 colon carcinomas and 9464D neuroblastoma were evaluated. Immune cell depletion and flow cytometric analysis were performed to determine the mechanisms of the antitumor effects. Results Tumors with volumes of 2,000 mm3 or larger were eradicated by CRI. Flow analyses of the tumors revealed reduction of T regulatory (Treg) cells and increase of CD8/Treg ratios following CRI. Rapid shrinkage of the treated tumors did not require T cells, whereas T cells were involved in the systemic effect against the distant tumors. Cured mice developed immunological memory. Conclusions These findings underscore that rationally designed combination immunotherapy regimens can be effective even against large, late-stage tumors.
Collapse
Affiliation(s)
| | - Noah W. Tsarovsky
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Mildred Felder
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Jen Zaborek
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, United States
| | - Sritha Moram
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Amy K. Erbe
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Alexander A. Pieper
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Dan V. Spiegelman
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Emily M. Cheng
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Cole M. Witt
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | | | - Zachary S. Morris
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
| | - Paul M. Sondel
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI, United States
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
9
|
Noh K, Choi H, Jo EH, Yoo W, Park KC. Role of SYT11 in human pan-cancer using comprehensive approaches. Eur J Med Res 2024; 29:338. [PMID: 38890718 PMCID: PMC11186215 DOI: 10.1186/s40001-024-01931-3] [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: 01/30/2024] [Accepted: 06/07/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Synaptotagmin 11 (SYT11) plays a pivotal role in neuronal vesicular trafficking and exocytosis. However, no independent prognostic studies have focused on various cancers. In this study, we aimed to summarize the clinical significance and molecular landscape of SYT11 in various tumor types. METHODS Using several available public databases, we investigated abnormal SYT11 expression in different tumor types and its potential clinical association with prognosis, methylation profiling, immune infiltration, gene enrichment analysis, and protein-protein interaction analysis, and identified common pathways. RESULTS TCGA and Genotype-Tissue Expression (GTEx) showed that SYT11 was widely expressed across tumor and corresponding normal tissues. Survival analysis showed that SYT11 expression correlated with the prognosis of seven cancer types. Additionally, SYT11 mRNA expression was not affected by promoter methylation, but regulated by certain miRNAs and associated with cancer patient prognosis. In vitro experiments further verified a negative correlation between the expression of SYT11 and miR-19a-3p in human colorectal, lung, and renal cancer cell lines. Moreover, aberrant SYT11 expression was significantly associated with immune infiltration. Pathway enrichment analysis revealed that the biological and molecular processes of SYT11 were related to clathrin-mediated endocytosis, Rho GTPase signaling, and cell motility-related functions. CONCLUSIONS Our results provide a clear understanding of the role of SYT11 in various cancer types and suggest that SYT11 may be of prognostic and clinical significance.
Collapse
Affiliation(s)
- Kyunghee Noh
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
- Department of Nanobiotechnology, University of Science and Technology (UST), Daejeon, 34141, Republic of Korea
| | - Hyunji Choi
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Eun-Hye Jo
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Wonbeak Yoo
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea.
| | - Kyung Chan Park
- Personalized Genomic Medicine Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea.
- Department of Functional Genomics, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea.
| |
Collapse
|
10
|
Xu S, Xing J, Li K, Qiao L, Zhang C, Ren Y, Liu Y. Pan-cancer analysis of DCTN2 and its tumour-promoting role in HCC by modulating the AKT pathway. J Cell Mol Med 2024; 28:e18450. [PMID: 38842133 PMCID: PMC11154834 DOI: 10.1111/jcmm.18450] [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: 02/01/2024] [Revised: 04/21/2024] [Accepted: 05/13/2024] [Indexed: 06/07/2024] Open
Abstract
Dynactin subunit 2 (DCTN2) has been reported to play a role in progression of several tumours; however, the involvement of DCTN2 in potential mechanism or the tumour immune microenvironment among various cancers still remains largely unknown. Therefore, the objective of this study was to comprehensively investigate the expression status and potential function of DCTN2 in various malignancies through different database, such as The Cancer Genome Atlas, the Genotype-Tissue Expression and Gene Expression Omnimus databases. We discovered that DCTN2 expression was high in many type of tumours tissues compared to adjacent non-tumour ones. High DCTN2 signified poor prognosis for patients with tumours. Additionally, Gene Set Enrichment Analysis (GSEA) analysis revealed that DCTN2 was positively correlated with oncogenic pathways, including cell cycle, tumour metastasis-related pathway, while it was negatively with anti-tumour immune signalling pathway, such as INF-γ response. More importantly, we elucidated the functional impact of DCTN2 on hepatocellular carcinoma (HCC) progression and its underlying mechanisms. DCTN2 expression was much higher in HCC tissues than in adjacent non-tumour tissues. Silencing DCTN2 dramatically suppressed the proliferative and metastasis capacities of tumour cell in vitro. Mechanistically, DCTN2 exerted tumour-promoting effects by modulating the AKT signalling pathway. DCTN2 knockdown in HCC cells inhibited AKT phosphorylation and its downstream targets as well. Rescue experiments revealed that the anti-tumour effects of DCTN2 knockdown were partially reversed upon AKT pathway activation. Overall, DCTN2 may be a potent biomarker signifying tumour prognosis and a promising therapeutic target for tumour treatment, particularly in HCC.
Collapse
Affiliation(s)
- Shuning Xu
- Department of Medical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Jiyuan Xing
- Department of Infectious DiseasesThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouChina
| | - Ke Li
- Department of Medical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Lei Qiao
- Department of Medical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Cheng Zhang
- Department of Medical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Yulin Ren
- Department of Medical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| | - Ying Liu
- Department of Medical OncologyThe Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer HospitalZhengzhouChina
| |
Collapse
|
11
|
Yang P, Shen G, Zhang H, Zhang C, Li J, Zhao F, Li Z, Liu Z, Wang M, Zhao J, Zhao Y. Incidence of thyroid dysfunction caused by immune checkpoint inhibitors combined with chemotherapy: A systematic review and meta-analysis. Int Immunopharmacol 2024; 133:111961. [PMID: 38608442 DOI: 10.1016/j.intimp.2024.111961] [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: 11/16/2023] [Revised: 03/05/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND The combination of immune checkpoint inhibitors (ICIs) and chemotherapy as a first-line treatment for triple-negative breast cancer (TNBC) has been associated with many adverse reactions. Thyroid dysfunction, the most common adverse reaction of the endocrine system, has also attracted significant attention. This study aimed to analyse the effect of ICIs combined with chemotherapy on thyroid function in patients with TNBC. METHODS As of November 4, 2023, we searched the PubMed, Web of Science, and Cochrane Library databases for clinical trials of ICIs combined with chemotherapy for the treatment of TNBC. The incidence of hypothyroidism and hyperthyroidism was calculated using a random-effects model. RESULTS In the final analysis, 3,226 patients from 19 studies were included. The total incidence of all-grade hypothyroidism induced by the combination of ICIs and chemotherapy in treating TNBC (12% (95% confidence intervals(CI): 0.10-0.15)) was higher than that of hyperthyroidism (5% (95% CI: 0.04-0.06)). Pembrolizumab combined with chemotherapy caused the highest incidence of all grades of hypothyroidism for 13% (95% CI: 0.05-0.06). Durvalumab combined with chemotherapy caused the highest incidence of all grades of hyperthyroidism, at 7% (95% CI: 0.03-0.11). ICIs combined with chemotherapy caused a higher incidence of all grades of hypothyroidism in advanced TNBC (15% (95% CI: 0.13-0.17)) than in early stage TNBC (10% (95% CI: 0.07-0.13)). CONCLUSION In TNBC, the incidence of hypothyroidism caused by the combination of ICIs and chemotherapy was significantly higher than that caused by hyperthyroidism. Pembrolizumab combined with chemotherapy resulted in the highest incidence of hypothyroidism. The incidence of hypothyroidism in patients with advanced TNBC was significantly higher than that in patients with early stage TNBC. In addition, ICIs combined with chemotherapy resulted in 16 out of 3,226 patients experiencing grade ≥ 3 thyroid dysfunction. Although the incidence of severe thyroid dysfunction is low, it requires attention. PROSPERO CRD42023477933.
Collapse
Affiliation(s)
- Ping Yang
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China.
| | - Guoshuang Shen
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China.
| | - Hengheng Zhang
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China.
| | - Chengrong Zhang
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China.
| | - Jinming Li
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China.
| | - Fuxing Zhao
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China.
| | - Zitao Li
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China.
| | - Zhen Liu
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China.
| | - Miaozhou Wang
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China.
| | - Jiuda Zhao
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China.
| | - Yi Zhao
- The Center of Breast Disease Diagnosis and Treatment of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining 810000, China.
| |
Collapse
|
12
|
Wang S, Li Y, Lin Y, Li J, Guo L, Wang H, Lin X, Liu Z, Zhang B, Liao Z, Zhang Z. Bioinformatics analysis and experimental verification of the cancer-promoting effect of DHODH in clear cell renal cell carcinoma. Sci Rep 2024; 14:11985. [PMID: 38796629 PMCID: PMC11127953 DOI: 10.1038/s41598-024-62738-0] [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: 03/25/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is a malignant tumor of the urinary system. To explore the potential mechanisms of DHODH in ccRCC, we analyzed its molecular characteristics using public databases. TCGA pan-cancer dataset was used to analyze DHODH expression in different cancer types and TCGA ccRCC dataset was used to assess differential expression, prognosis correlation, immune infiltration, single-gene, and functional enrichment due to DHODH. The GSCALite and CellMiner databases were employed to explore drugs and perform molecular docking analysis with DHODH. Protein-protein interaction networks and ceRNA regulatory networks of DHODH were constructed using multiple databases. The effect of DHODH on ccRCC was confirmed in vitro. DHODH was highly expressed in ccRCC. Immune infiltration analysis revealed that DHODH may be involved in regulating the infiltration of immunosuppressive cells such as Tregs. Notably, DHODH influenced ccRCC progression by forming regulatory networks with molecules, such as hsa-miR-26b-5p and UMPS and significantly enhanced the malignant characteristics of ccRCC cells. Several drugs, such as lapatinib, silmitasertib, itraconazole, and dasatinib, were sensitive to DHODH expression and exhibited strong molecular binding with it. Thus, DHODH may promote ccRCC progression and is a candidate effective therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Songsong Wang
- Department of Urology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
- School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Yan Li
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China
| | - Yilong Lin
- School of Medicine, Xiamen University, Xiamen, 361000, China
- The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Junting Li
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350000, China
| | - Lang Guo
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China
| | - Haoyu Wang
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China
| | - Xinyuan Lin
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350000, China
| | - Ziming Liu
- Department of Urology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China.
- School of Medicine, Xiamen University, Xiamen, 361000, China.
| | - Bingqi Zhang
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China.
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China.
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China.
| | - Zhengming Liao
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China.
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China.
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China.
| | - Zhongmin Zhang
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China.
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China.
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China.
| |
Collapse
|
13
|
Jin SX, Liu BN, Ji HJ, Wu JR, Li BL, Gao XL, Li N, Zheng ZD, Du C. Serum cytokines and creatinine/cystatin C ratio as prognostic biomarkers in advanced cancer patients treated with anti-PD-1/PD-L1 therapy. Support Care Cancer 2024; 32:370. [PMID: 38776028 PMCID: PMC11111483 DOI: 10.1007/s00520-024-08525-z] [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: 02/11/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE Immune checkpoint inhibitors (ICIs), specifically targeting the programmed cell death protein-1 or its ligand (PD-1/PD-L1), have been extensively used in the treatment of a spectrum of malignancies, although the predictive biomarkers remain to be elucidated. This study aims to investigate the association between baseline circulating levels of cytokines and the creatinine/cystatin C ratio (CCR) with the treatment outcomes of ICIs in patients with advanced cancer. METHODS The pre-treatment circulating levels of 10 cytokines (PD-L1, CTLA4, CXCL10, LAG3, HGF, CCL2, MIG, GRANB, IL-18, and IL-6) were measured via automated capillary-based immunoassay platform in the serum of 65 advanced cancer patients treated with anti-PD-1/PD-L1-based systemic therapy and 10 healthy volunteers. The levels of cytokines and CCR were quantified and categorized into high and low groups based on the median value. The associations of serum cytokines and CCR with response to treatment, survival, and immune-related adverse events were assessed. RESULTS Elevated circulating levels of 6 cytokines (PD-L1, CXCL10, HGF, CCL2, MIG, and IL-6) were observed in cancer patients compared with that in healthy volunteers. The correlation coefficients between cytokines, CCR and nutritional risk index were also calculated. In the cancer cohort (N = 65), low circulating HGF (P = 0.023, P = 0.029), low IL-6 (P = 0.002, P < 0.001), and high CCR (P = 0.031, P = 0.008) were associated with significantly improved progression-free survival (PFS) and overall survival (OS). Multi-variable COX analyses adjusted for clinicopathological factors revealed that low HGF, low IL-6, and high CCR were independent favorable prognostic factors for PFS (P = 0.028, P = 0.010, and P = 0.015, respectively) and OS (P = 0.043, P = 0.003, and P = 0.026, respectively). Grade 2 irAEs occurred more frequently in patients with low levels of circulating CCL2 and LAG3. CONCLUSIONS Pre-treatment circulating levels of serum IL-6, HGF, and CCR may serve as independent predictive and prognostic biomarkers in advanced cancer patients treated with ICIs-based systemic therapy. These findings might help to identify potential patients who would benefit from these therapies.
Collapse
Affiliation(s)
- Shan-Xiu Jin
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, China
| | - Bo-Na Liu
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, China
| | - Hong-Juan Ji
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, China
| | - Jing-Ran Wu
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, China
| | - Bao-Lei Li
- Department of Oncology, Anshan Tumor Hospital, Anshan, China
| | - Xiao-Li Gao
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, China
| | - Na Li
- Department of Gynaecology and Obstetrics, The First Hospital of Jilin University, Jilin, China.
| | - Zhen-Dong Zheng
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, China.
| | - Cheng Du
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, China.
| |
Collapse
|
14
|
Rydén V, El-Naggar AI, Koliadi A, Ladjevardi CO, Digkas E, Valachis A, Ullenhag GJ. The role of dacarbazine and temozolomide therapy after treatment with immune checkpoint inhibitors in malignant melanoma patients: A case series and meta-analysis. Pigment Cell Melanoma Res 2024; 37:352-362. [PMID: 38158376 DOI: 10.1111/pcmr.13156] [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: 09/13/2023] [Revised: 11/17/2023] [Accepted: 12/12/2023] [Indexed: 01/03/2024]
Abstract
Dacarbazine (DTIC) and its oral counterpart temozolomide (TMZ) have been the most used agents in advanced malignant melanoma (MM) patients and they are still used routinely. The preferred first line treatment, immune checkpoint inhibitors (CPIs) might shape the tumor and the tumor microenvironment, possibly affecting the response to subsequent therapies. The aim of this study was to investigate the treatment effect of DTIC/TMZ in MM patients after CPI therapy in a consecutive patient cohort and through systematic literature review and meta-analysis. Thirty-five patients with advanced MM treated with DTIC/TMZ after previous CPI therapy in three Swedish regions between 2017 and 2021 were recognized and seven case series studies were identified through systematic database review. Pooled data from all 345 patients showed a median real-world progression-free survival (rwPFS) of 1.9 months and overall survival (OS) of 6.0 months. Three of these studies were included in a meta-analysis comparing DTIC/TMZ after CPI treatment, versus no previous immunotherapy, showing no statistically significant differences in rwPFS or OS but higher real-world response rate to chemotherapy for the prior-CPI treated group (Odds Ratio: 2.24; 95% Confidence Interval: 1.04-4.86). The current study supports consideration of DTIC/TMZ in later line of treatment in the immunotherapy era.
Collapse
Affiliation(s)
- Viktoria Rydén
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Ali Inan El-Naggar
- Department of Oncology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Department of Oncology, Örebro University Hospital, Örebro, Sweden
| | - Anthoula Koliadi
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Cecilia Olsson Ladjevardi
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| | - Evangelos Digkas
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
- Department of Oncology, Eskilstuna, Sweden
| | - Antonios Valachis
- Department of Oncology, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
- Department of Oncology, Örebro University Hospital, Örebro, Sweden
| | - Gustav J Ullenhag
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Oncology, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
15
|
Saito-Koyama R, Tamai K, Yasuda J, Okamura Y, Yamazaki Y, Inoue C, Miki Y, Abe J, Oishi H, Sato I, Sasano H. Morphometric analysis of nuclear shape irregularity as a novel predictor of programmed death-ligand 1 expression in lung squamous cell carcinoma. Virchows Arch 2024; 484:609-620. [PMID: 37171482 DOI: 10.1007/s00428-023-03548-z] [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: 12/06/2022] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 05/13/2023]
Abstract
Immune checkpoint inhibitor (ICI) therapy has been established as one of the key treatment strategies for lung squamous cell carcinoma (LUSQ). The status of programmed death-ligand 1 (PD-L1) in tumor cells and/or immune cells using immunohistochemistry has been primarily used as a surrogate marker for determining ICI treatment; however, when the tissues to be examined are small, false-negative results could be unavoidable due to the heterogeneity of PD-L1 immunoreactivity. To overcome this practical limitation, we attempted to explore the status of nuclear atypia evaluated using morphometry as a potential predictor of PD-L1 status in LUSQ. We correlated the parameters related to nuclear atypia with PD-L1 status using two different cohorts of LUSQ patients (95 cases from The Cancer Genome Atlas database and 30 cases from the Miyagi Cancer Center). Furthermore, we studied the gene mutation status to elucidate the genetic profile of PD-L1 predictable cases. The results revealed that nuclear atypia, especially morphometric parameters related to nuclear shape irregularity, including aspect ratio, circularity, roundness, and solidity, were all significantly associated with PD-L1 status. Additionally, LUSQ cases with high PD-L1 expression and pronounced nuclear atypia were significantly associated with C10orf71 and COL14A1 mutations compared with those with low PD-L1 expression and mild nuclear atypia. We demonstrated for the first time that nuclear shape irregularity could represent a novel predictor of PD-L1 expression in LUSQ. Including the morphometric parameters related to nuclear atypia in conjunction with PD-L1 status could help determine an effective ICI therapeutic strategy; however, further investigation is required.
Collapse
Affiliation(s)
- Ryoko Saito-Koyama
- Department of Pathology, Tohoku University School of Medicine, Miyagi, Japan.
- Department of Pathology, National Hospital Organization, Sendai Medical Center, 2-11-12 Miyagino, Miyagino-ku, Sendai, Miyagi, 983-8520, Japan.
| | - Keiichi Tamai
- Division of Cancer Stem Cell, Miyagi Cancer Center Research Institute, Miyagi, Japan
| | - Jun Yasuda
- Division of Molecular and Cellular Oncology, Miyagi Cancer Center Research Institute, Miyagi, Japan
| | - Yasunobu Okamura
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Miyagi, Japan
| | - Yuto Yamazaki
- Department of Pathology, Tohoku University School of Medicine, Miyagi, Japan
| | - Chihiro Inoue
- Department of Pathology, Tohoku University School of Medicine, Miyagi, Japan
| | - Yasuhiro Miki
- Department of Pathology, Tohoku University School of Medicine, Miyagi, Japan
- Faculty of Medical Science & Welfare, Tohoku Bunka Gakuen University, Miyagi, Japan
| | - Jiro Abe
- Division of Thoracic Surgery, Miyagi Cancer Center, Miyagi, Japan
| | - Hisashi Oishi
- Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Miyagi, Japan
| | - Ikuro Sato
- Division of Pathology, Miyagi Cancer Center, Miyagi, Japan
| | - Hironobu Sasano
- Department of Pathology, Tohoku University School of Medicine, Miyagi, Japan
| |
Collapse
|
16
|
Lan HR, Chen M, Yao SY, Chen JX, Jin KT. Novel immunotherapies for breast cancer: Focus on 2023 findings. Int Immunopharmacol 2024; 128:111549. [PMID: 38266449 DOI: 10.1016/j.intimp.2024.111549] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
Immunotherapy has emerged as a revolutionary approach in cancer therapy, and recent advancements hold significant promise for breast cancer (BCa) management. Employing the patient's immune system to combat BCa has become a focal point in immunotherapeutic investigations. Strategies such as immune checkpoint inhibitors (ICIs), adoptive cell transfer (ACT), and targeting the tumor microenvironment (TME) have disclosed encouraging clinical outcomes. ICIs, particularly programmed cell death protein 1 (PD-1)/PD-L1 inhibitors, exhibit efficacy in specific BCa subtypes, including triple-negative BCa (TNBC) and human epidermal growth factor receptor 2 (HER2)-positive cancers. ACT approaches, including tumor-infiltrating lymphocytes (TILs) and chimeric antigen receptor (CAR) T-cell therapy, showed promising clinical outcomes in enhancing tumor recognition and elimination. Targeting the TME through immune agonists and oncolytic viruses signifies a burgeoning field of research. While challenges persist in patient selection, resistance mechanisms, and combination therapy optimization, these novel immunotherapies hold transformative potential for BCa treatment. Continued research and clinical trials are imperative to refine and implement these innovative approaches, paving the way for improved outcomes and revolutionizing the management of BCa. This review provides a concise overview of the latest immunotherapies (2023 studies) in BCa, highlighting their potential and current status.
Collapse
Affiliation(s)
- Huan-Rong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, China
| | - Min Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Shi-Ya Yao
- Department of Gastrointestinal, Colorectal and Anal Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China
| | - Jun-Xia Chen
- Department of Gynecology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, China.
| | - Ke-Tao Jin
- Department of Gastrointestinal, Colorectal and Anal Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China.
| |
Collapse
|
17
|
Shin S, Moon J, Oum C, Kim S, Cho SI, Lim Y, Ock CY, Shin S. Discontinuation risk from adverse events: immunotherapy alone vs. combined with chemotherapy: a systematic review and network meta-analysis. BMC Cancer 2024; 24:152. [PMID: 38291376 PMCID: PMC10825980 DOI: 10.1186/s12885-024-11897-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/19/2024] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND While immunotherapy combined with chemotherapy (Chemo-IO) is generally recognized for providing superior outcomes compared to monotherapy (mono-IO), it is associated with a higher incidence of treatment-related adverse events (TRAEs), which may lead to treatment discontinuation. In this study, we compared the rates of treatment discontinuation between mono-IO and Chemo-IO as first-line treatments for various solid tumors. METHODS We systematically reviewed clinical trials from databases (PubMed, Embase, Cochrane Library, and an additional source) published from January 1, 2018, to July 10, 2023. We included phase III randomized controlled trials (RCTs) that utilized immunotherapy agents in at least one arm as first-line treatments for a variety of solid tumors. Data extraction followed the Preferred Reporting Items for Systematic Reviews (PRISMA) extension statement for network meta-analysis. A random effects model was used for the network meta-analysis, with the risk of bias assessed using the Cochrane risk-of-bias tool II. The primary outcomes encompassed treatment discontinuation rates due to TRAEs among patients who underwent immunotherapy, either alone or combined with chemotherapy, for various solid tumors. Pooled relative risks (RRs) with 95% confidence intervals (CIs) were calculated to compare between treatment groups. RESULTS From 29 RCTs, a total of 21,677 patients and 5 types of treatment were analyzed. Compared to mono-IO, Chemo-IO showed a significantly higher rate of discontinuation due to TRAEs (RR 2.68, 95% CI 1.98-3.63). Subgroup analysis for non-small cell lung cancer (NSCLC) patients also exhibited a greater risk of discontinuation due to TRAEs with Chemo-IO compared to mono-IO (RR 2.93, 95% CI 1.67-5.14). Additional analyses evaluating discontinuation rates due to either treatment emergent adverse events (TEAEs) or AEs regardless of causality (any AEs) consistently revealed an elevated risk associated with Chemo-IO. CONCLUSIONS Chemo-IO was associated with an elevated risk of treatment discontinuation not only due to TRAEs but also any AEs or TEAEs. Given that the treatment duration can impact clinical outcomes, a subset of patients might benefit more from mono-IO than combination therapy. Further research is imperative to identify and characterize this subset.
Collapse
Affiliation(s)
- Sangwon Shin
- Lunit, 4th to 8th floors, 374, Gangnam-daero, Gangnam-gu, Seoul, Republic of Korea
| | - Jimin Moon
- Lunit, 4th to 8th floors, 374, Gangnam-daero, Gangnam-gu, Seoul, Republic of Korea
| | - Chiyoon Oum
- Lunit, 4th to 8th floors, 374, Gangnam-daero, Gangnam-gu, Seoul, Republic of Korea
| | - Seulki Kim
- Lunit, 4th to 8th floors, 374, Gangnam-daero, Gangnam-gu, Seoul, Republic of Korea
| | - Soo Ick Cho
- Lunit, 4th to 8th floors, 374, Gangnam-daero, Gangnam-gu, Seoul, Republic of Korea
| | - Yoojoo Lim
- Lunit, 4th to 8th floors, 374, Gangnam-daero, Gangnam-gu, Seoul, Republic of Korea
| | - Chan-Young Ock
- Lunit, 4th to 8th floors, 374, Gangnam-daero, Gangnam-gu, Seoul, Republic of Korea
| | - Seunghwan Shin
- Lunit, 4th to 8th floors, 374, Gangnam-daero, Gangnam-gu, Seoul, Republic of Korea.
| |
Collapse
|
18
|
Liu R, Liang Q, Luo J, Li Y, Zhang X, Fan K, Du J. Ferritin-Based Nanocomposite Hydrogel Promotes Tumor Penetration and Enhances Cancer Chemoimmunotherapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305217. [PMID: 38029345 PMCID: PMC10797422 DOI: 10.1002/advs.202305217] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/19/2023] [Indexed: 12/01/2023]
Abstract
Hydrogels are prevailing drug delivery depots to improve antitumor efficacy and reduce systemic toxicity. However, the application of conventional free drug-loaded hydrogel is hindered by poor drug penetration in solid tumors. Here, an injectable ferritin-based nanocomposite hydrogel is constructed to facilitate tumor penetration and improve cancer chemoimmunotherapy. Specifically, doxorubicin-loaded human ferritin (Dox@HFn) and oxidized dextran (Dex-CHO) are used to construct the injectable hydrogel (Dox@HFn Gel) through the formation of pH-sensitive Schiff-base bonds. After peritumoral injection, the Dox@HFn Gel is retained locally for up to three weeks, and released intact Dox@HFn gradually, which can not only facilitate tumor penetration through active transcytosis but also induce immunogenic cell death (ICD) to tumor cells to generate an antitumor immune response. Combining with anti-programmed death-1 antibody (αPD-1), Dox@HFn Gel induces remarkable regression of orthotopic 4T1 breast tumors, further elicits a strong systemic anti-tumor immune response to effectively suppress tumor recurrence and lung metastasis of 4T1 tumors after surgical resection. Besides, the combination of Dox@HFn GelL with anti-CD47 antibody (αCD47) inhibits postsurgical tumor recurrence of aggressive orthotopic glioblastoma tumor model and significantly extends mice survival. This work sheds light on the construction of local hydrogels to potentiate antitumor immune response for improved cancer therapy.
Collapse
Affiliation(s)
- Rong Liu
- School of MedicineSouth China University of TechnologyGuangzhou510006China
| | - Qian Liang
- CAS Engineering Laboratory for NanozymeNational Laboratory of BiomacromoleculesInstitute of BiophysicsChinese Academy of SciencesBeijing100101China
| | - Jia‐Qi Luo
- School of MedicineSouth China University of TechnologyGuangzhou510006China
| | - Yu‐Xuan Li
- School of Biomedical Sciences and EngineeringGuangzhou International CampusSouth China University of TechnologyGuangzhou511442China
| | - Xin Zhang
- School of MedicineSouth China University of TechnologyGuangzhou510006China
| | - Kelong Fan
- CAS Engineering Laboratory for NanozymeNational Laboratory of BiomacromoleculesInstitute of BiophysicsChinese Academy of SciencesBeijing100101China
- University of Chinese Academy of SciencesBeijing101408China
- Nanozyme Medical CenterSchool of Basic Medical SciencesZhengzhou UniversityZhengzhou450052China
| | - Jin‐Zhi Du
- School of MedicineSouth China University of TechnologyGuangzhou510006China
- National Engineering Research Center for Tissue Restoration and ReconstructionSouth China University of TechnologyGuangzhou510006China
- Key Laboratory of Biomedical Materials and EngineeringMinistry of EducationGuangdong Provincial Key Laboratory of Biomedical EngineeringSouth China University of TechnologyGuangzhou510006China
| |
Collapse
|
19
|
Chen J, Cui M, He L, Mu Y, Hu N, Guan X. Engineered elastin-like polypeptide-based hydrogel delivering chemotherapeutics and PD-L1 antibodies for potentiated cancer immunotherapy. J Mater Chem B 2023; 11:10355-10361. [PMID: 37817648 DOI: 10.1039/d3tb01974h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have effectively eradicated advanced tumors by inducing durable and systematic antitumor immune responses. However, when used as a standalone treatment, ICIs typically exhibit a low response rate in many cancers. In this study, we engineered an in situ-formed gel depot using elastin-like polypeptides (ELPs) to efficiently deliver PD-L1 antibodies (aPD-L1) and gemcitabine (GEM) for enhanced immunotherapy in melanoma. Sustainably released chemotherapeutics from gel depots could kill melanoma cells and promote PD-L1 upregulation in tumor cells. Moreover, aPD-L1/GEM-encapsulated ELP hydrogel promoted a 3.0-fold increase of tumor-infiltrated CD8+ T cells and 60% Tregs depletion compared with PBS group, eliciting a robust antitumor immune response for immunotherapy in melanoma mouse models. This research highlights the promising potential of ELP-based hydrogels in delivering ICIs and chemotherapeutic agents for potentiated cancer immunotherapy.
Collapse
Affiliation(s)
- Jinguang Chen
- Department of Dermatology, Taizhou Central Hospital (Taizhou University Hospital), Taizhou University, Taizhou 318001, P. R. China
| | - Meiying Cui
- Department of Clinical Laboratory, Affiliated Hospital of Jining Medical College, Jining 272007, China
- Department of Basic Medical Science, Medical School, Taizhou University, Taizhou 318001, P. R. China.
| | - Lianping He
- Department of Basic Medical Science, Medical School, Taizhou University, Taizhou 318001, P. R. China.
| | - Yeteng Mu
- Department of Basic Medical Science, Medical School, Taizhou University, Taizhou 318001, P. R. China.
| | - Nannan Hu
- Department of Basic Medical Science, Medical School, Taizhou University, Taizhou 318001, P. R. China.
| | - Xingang Guan
- Department of Basic Medical Science, Medical School, Taizhou University, Taizhou 318001, P. R. China.
| |
Collapse
|
20
|
Baas IO, Westermann AM, You B, Bolze PA, Seckl M, Ghorani E. Immunotherapy for Gestational Trophoblastic Neoplasia: A New Paradigm. Gynecol Obstet Invest 2023; 89:230-238. [PMID: 37703867 PMCID: PMC11152029 DOI: 10.1159/000533972] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/27/2023] [Indexed: 09/15/2023]
Abstract
BACKGROUND Immune checkpoint immunotherapy (CPI) targeting programmed cell death 1 (PD-1)/ligand (PD-L1) has been shown to be an effective treatment for gestational trophoblastic neoplasia (GTN). This includes those with multidrug resistance, ultra-high-risk disease, and epithelioid trophoblastic tumour/placental site trophoblastic tumour subtypes that are inherently chemotherapy resistant, but there is also emerging evidence in low-risk disease. OBJECTIVES We set out to generate an overview of the current data supporting the use of CPI for GTN in both high-risk and low-risk disease and to consider future research goals and directions in order to implement CPI in current treatment guidelines. METHODS We identified and reviewed the published data on the use of CPI agents in GTN. OUTCOME 133 patients were identified who had been treated with CPI for GTN with pembrolizumab (23), avelumab (22), camrelizumab (57), toripalimab (15), or other anti-PD-1 agents (16), of whom 118 had high-risk diseases, relapse or multi-drug resistant disease, and 15 low-risk diseases. Overall 85 patients achieved complete remission, 77 (of 118) with high-risk disease, and 8 (of 15) with low-risk disease. 1 patient with complete remission in the high-risk group developed a relapse 22 months after anti-PD-1 treatment had been stopped. Treatment was generally well tolerated across studies. CONCLUSIONS AND OUTLOOK The majority of high-risk patients (77/118) treated with CPI are cured and this is particularly relevant amongst those with chemotherapy resistant disease who otherwise have very limited treatment options. Priorities for future research include determining whether these agents have a role earlier in the disease course, the utility of combination with chemotherapy, and effects on future fertility. Treatment availability remains a concern due to the high price of these agents.
Collapse
Affiliation(s)
- Inge O Baas
- Department of Medical Oncology, University Medical Centre Utrecht, Utrecht, The Netherlands,
| | - Anneke M Westermann
- Department of Medical Oncology, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, The Netherlands
| | - Benoit You
- Université Lyon 1, Hospices Civils De Lyon, Centre Français De Référence Des Maladies Trophoblastiques, Hôpital Lyon Sud, CITOHL, EA 3738 CICLY, Univ Lyon 1, Lyon, France
| | - Pierre-Adrien Bolze
- Université Lyon 1, Hospices Civils De Lyon, Centre Français De Référence Des Maladies Trophoblastiques, Hôpital Lyon Sud, CITOHL, EA 3738 CICLY, Univ Lyon 1, Lyon, France
| | - Michael Seckl
- Department of Medical Oncology, Gestational Trophoblastic Disease Centre, Imperial College Healthcare NHS Trust, Charing Cross Hospital, London, UK
| | - Ehsan Ghorani
- Department of Medical Oncology, Gestational Trophoblastic Disease Centre, Imperial College Healthcare NHS Trust, Charing Cross Hospital, London, UK
| |
Collapse
|
21
|
Lobo CS, Mendes MIP, Pereira DA, Gomes-da-Silva LC, Arnaut LG. Photodynamic therapy changes tumour immunogenicity and promotes immune-checkpoint blockade response, particularly when combined with micromechanical priming. Sci Rep 2023; 13:11667. [PMID: 37468749 DOI: 10.1038/s41598-023-38862-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 07/16/2023] [Indexed: 07/21/2023] Open
Abstract
Photodynamic therapy (PDT) with redaporfin stimulates colon carcinoma (CT26), breast (4T1) and melanoma (B16F10) cells to display high levels of CD80 molecules on their surfaces. CD80 overexpression amplifies immunogenicity because it increases same cell (cis) CD80:PD-L1 interactions, which (i) disrupt binding of T-cells PD-1 inhibitory receptors with their ligands (PD-L1) in tumour cells, and (ii) inhibit CTLA-4 inhibitory receptors binding to CD80 in tumour cells. In some cancer cells, redaporfin-PDT also increases CTLA-4 and PD-L1 expressions and virtuous combinations between PDT and immune-checkpoint blockers (ICB) depend on CD80/PD-L1 or CD80/CTLA-4 tumour overexpression ratios post-PDT. This was confirmed using anti-CTLA-4 + PDT combinations to increase survival of mice bearing CT26 tumours, and to regress lung metastases observed with bioluminescence in mice with orthotopic 4T1 tumours. However, the primary 4T1 responded poorly to treatments. Photoacoustic imaging revealed low infiltration of redaporfin in the tumour. Priming the primary tumour with high-intensity (~ 60 bar) photoacoustic waves generated with nanosecond-pulsed lasers and light-to-pressure transducers improved the response of 4T1 tumours to PDT. Penetration-resistant tumours require a combination of approaches to respond to treatments: tumour priming to facilitate drug infiltration, PDT for a strong local effect and a change in immunogenicity, and immunotherapy for a systemic effect.
Collapse
Affiliation(s)
- Catarina S Lobo
- CQC, Chemistry Department, University of Coimbra, 3004-535, Coimbra, Portugal
| | - Maria Inês P Mendes
- CQC, Chemistry Department, University of Coimbra, 3004-535, Coimbra, Portugal
| | - Diogo A Pereira
- CQC, Chemistry Department, University of Coimbra, 3004-535, Coimbra, Portugal
| | | | - Luis G Arnaut
- CQC, Chemistry Department, University of Coimbra, 3004-535, Coimbra, Portugal.
| |
Collapse
|
22
|
Silva-Pilipich N, Covo-Vergara Á, Vanrell L, Smerdou C. Checkpoint blockade meets gene therapy: Opportunities to improve response and reduce toxicity. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 379:43-86. [PMID: 37541727 DOI: 10.1016/bs.ircmb.2023.05.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/06/2023]
Abstract
Immune checkpoint inhibitors (ICIs) based on monoclonal antibodies represent a breakthrough for the treatment of cancer. However, their efficacy varies among tumor types and patients, and they can lead to adverse effects due to on-target/off-tumor activity, since they are administered systemically at high doses. An alternative and attractive approach for the delivery of ICIs is the use of gene therapy vectors able to express them in vivo. This review focuses on the most recent studies using viral vectors able to express ICIs locally or systemically in preclinical models of cancer. These vectors include non-replicating viruses, oncolytic viruses able to propagate specifically in tumor cells and destroy them, and self-amplifying RNA vectors, armed with different formats of antibodies against immune checkpoints. Non-replicating vectors usually lead to long-term ICI expression, potentially eliminating the need for repeated administration. Vectors with replication capacity, although they have a shorter window of expression, can induce inflammation which enhances the antitumor effect. Finally, these engineered vectors can be used in combination with other immunostimulatory molecules or with CAR-T cells, further boosting the antitumor immune responses.
Collapse
Affiliation(s)
- Noelia Silva-Pilipich
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IdISNA), and CCUN, Pamplona, Spain.
| | - Ángela Covo-Vergara
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IdISNA), and CCUN, Pamplona, Spain
| | - Lucía Vanrell
- Facultad de Ingeniería, Universidad ORT Uruguay, Montevideo, Uruguay; Nanogrow Biotech, Montevideo, Uruguay
| | - Cristian Smerdou
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IdISNA), and CCUN, Pamplona, Spain.
| |
Collapse
|
23
|
Desai NV, Tan AR. Targeted Therapies and the Evolving Standard of Care for Triple-Negative and Germline BRCA1/ 2-Mutated Breast Cancers in the High-Risk, Early-Stage Setting. JCO Precis Oncol 2023; 7:e2200446. [PMID: 37163718 DOI: 10.1200/po.22.00446] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023] Open
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs) and poly (ADP-ribose) polymerase (PARP) inhibitors have transformed the treatment landscape of metastatic triple-negative breast cancer (TNBC). Trial results have demonstrated the clinical benefit of these targeted agents in the advanced TNBC setting and have led to their evaluation in the treatment of high-risk, early-stage TNBC and BRCA-mutated breast cancer. We provide a summary of the results that have led to the establishment of the ICI pembrolizumab and the PARP inhibitor olaparib as new standards of care. METHODS Using PubMed, we searched for original articles published in English between 2017 and 2022. Search terms included triple-negative breast cancer, adjuvant, neoadjuvant, immunotherapy, and PARP inhibitors. RESULTS Two targeted therapies have been approved by the US Food and Drug Administration for the treatment of TNBC and BRCA-mutated breast cancers in the high-risk, early-stage setting on the basis of clinical trial results demonstrating improved clinical outcomes. For high-risk, early-stage TNBC, pembrolizumab was approved as neoadjuvant therapy in combination with chemotherapy and as a single agent for continued treatment after surgery; this approval was based on results of the KEYNOTE-522 trial. Olaparib was approved for the adjuvant treatment of patients with high-risk, early-stage human epidermal growth factor receptor type 2 (HER2)-negative breast cancer with germline BRCA1/2 mutations who have been previously treated with neoadjuvant or adjuvant chemotherapy on the basis of the OlympiA trial results. CONCLUSION Clinical trial results demonstrate the pronounced clinical benefits of pembrolizumab combined with chemotherapy for high-risk, early-stage TNBC and adjuvant olaparib for high-risk, early-stage HER2-negative BRCA1/2-mutated breast cancer.
Collapse
Affiliation(s)
- Neelam V Desai
- Department of Solid Tumor Oncology and Investigational Therapeutics, Levine Cancer Institute, Atrium Health, Charlotte, NC
| | - Antoinette R Tan
- Department of Solid Tumor Oncology and Investigational Therapeutics, Levine Cancer Institute, Atrium Health, Charlotte, NC
| |
Collapse
|
24
|
Kiousi DE, Kouroutzidou AZ, Neanidis K, Karavanis E, Matthaios D, Pappa A, Galanis A. The Role of the Gut Microbiome in Cancer Immunotherapy: Current Knowledge and Future Directions. Cancers (Basel) 2023; 15:cancers15072101. [PMID: 37046762 PMCID: PMC10093606 DOI: 10.3390/cancers15072101] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Cancer immunotherapy is a treatment modality that aims to stimulate the anti-tumor immunity of the host to elicit favorable clinical outcomes. Immune checkpoint inhibitors (ICIs) gained traction due to the lasting effects and better tolerance in patients carrying solid tumors in comparison to conventional treatment. However, a significant portion of patients may present primary or acquired resistance (non-responders), and thus, they may have limited therapeutic outcomes. Resistance to ICIs can be derived from host-related, tumor-intrinsic, or environmental factors. Recent studies suggest a correlation of gut microbiota with resistance and response to immunotherapy as well as with the incidence of adverse events. Currently, preclinical and clinical studies aim to elucidate the unique microbial signatures related to ICI response and anti-tumor immunity, employing metagenomics and/or multi-omics. Decoding this complex relationship can provide the basis for manipulating the malleable structure of the gut microbiota to enhance therapeutic success. Here, we delve into the factors affecting resistance to ICIs, focusing on the intricate gut microbiome–immunity interplay. Additionally, we review clinical studies and discuss future trends and directions in this promising field.
Collapse
Affiliation(s)
- Despoina E. Kiousi
- Department of Molecular Biology and Genetics, Faculty of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Antonia Z. Kouroutzidou
- Department of Molecular Biology and Genetics, Faculty of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Konstantinos Neanidis
- Oncology Department, 424 General Military Training Hospital, 56429 Thessaloniki, Greece
| | - Emmanuel Karavanis
- Oncology Department, 424 General Military Training Hospital, 56429 Thessaloniki, Greece
| | | | - Aglaia Pappa
- Department of Molecular Biology and Genetics, Faculty of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Alex Galanis
- Department of Molecular Biology and Genetics, Faculty of Health Sciences, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| |
Collapse
|
25
|
Hennah L, Seckl M, Ghorani E. Novel approaches to managing gestational trophoblastic tumors in the age of immunotherapy. Int J Gynecol Cancer 2023; 33:414-419. [PMID: 36878565 DOI: 10.1136/ijgc-2022-003771] [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] [Indexed: 03/08/2023] Open
Abstract
The discovery that anti-programmed death-1 antibody (anti-PD-1) immunotherapy can cure patients with multidrug-resistant gestational trophoblastic neoplasia provides a new powerful and low toxicity treatment. This heralds an era within which the majority of patients, including those with previously difficult to treat disease, can expect to achieve long-term remission. This development should prompt a rethink of how patients with this rare disease are managed, focusing on maximizing cure rate with minimal exposure to toxic chemotherapy.
Collapse
Affiliation(s)
- Lindsay Hennah
- Department of Medical Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - Michael Seckl
- Department of Medical Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - Ehsan Ghorani
- Department of Medical Oncology, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
26
|
Abstract
Immunotherapy has revolutionized the treatment of patients with cancer. However, promoting antitumour immunity in patients with tumours that are resistant to these therapies remains a challenge. Thermal therapies provide a promising immune-adjuvant strategy for use with immunotherapy, mostly owing to the capacity to reprogramme the tumour microenvironment through induction of immunogenic cell death, which also promotes the recruitment of endogenous immune cells. Thus, thermal immunotherapeutic strategies for various cancers are an area of considerable research interest. In this Review, we describe the role of the various thermal therapies and provide an update on attempts to combine these with immunotherapies in clinical trials. We also provide an overview of the preclinical development of various thermal immuno-nanomedicines, which are capable of combining thermal therapies with various immunotherapy strategies in a single therapeutic platform. Finally, we discuss the challenges associated with the clinical translation of thermal immuno-nanomedicines and emphasize the importance of multidisciplinary and inter-professional collaboration to facilitate the optimal translation of this technology from bench to bedside.
Collapse
|
27
|
Mei L, Zhang Z, Li X, Yang Y, Qi R. Metabolomics profiling in prediction of chemo-immunotherapy efficiency in advanced non-small cell lung cancer. Front Oncol 2023; 12:1025046. [PMID: 36733356 PMCID: PMC9887290 DOI: 10.3389/fonc.2022.1025046] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 12/28/2022] [Indexed: 01/18/2023] Open
Abstract
Background To explore potential metabolomics biomarker in predicting the efficiency of the chemo-immunotherapy in patients with advanced non-small cell lung cancer (NSCLC). Methods A total of 83 eligible patients were assigned to receive chemo-immunotherapy. Serum samples were prospectively collected before the treatment to perform metabolomics profiling analyses under the application of gas chromatography mass spectrometry (GC-MS). The key metabolites were identified using projection to latent structures discriminant analysis (PLS-DA). The key metabolites were used for predicting the chemo-immunotherapy efficiency in advanced NSCLC patients. Results Seven metabolites including pyruvate, threonine, alanine, urea, oxalate, elaidic acid and glutamate were identified as the key metabolites to the chemo-immunotherapy response. The receiver operating characteristic curves (AUC) were 0.79 (95% CI: 0.69-0.90), 0.60 (95% CI: 0.48-0.73), 0.69 (95% CI: 0.57-0.80), 0.63 (95% CI: 0.51-0.75), 0.60 (95% CI: 0.48-0.72), 0.56 (95% CI: 0.43-0.67), and 0.67 (95% CI: 0.55-0.80) for the key metabolites, respectively. A binary logistic regression was used to construct a combined biomarker model to improve the discriminating efficiency. The AUC was 0.86 (95% CI: 0.77-0.94) for the combined biomarker model. Pathway analyses showed that urea cycle, glucose-alanine cycle, glycine and serine metabolism, alanine metabolism, and glutamate metabolism were the key metabolic pathway to the chemo-immunotherapy response in patients with advanced NSCLC. Conclusion Metabolomics analyses of key metabolites and pathways revealed that GC-MS could be used to predict the efficiency of chemo-immunotherapy. Pyruvate, threonine, alanine, urea, oxalate, elaidic acid and glutamate played a central role in the metabolic of PD patients with advanced NSCLC.
Collapse
Affiliation(s)
- Lihong Mei
- Department of Dermatology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Zhihua Zhang
- Department of Echocardiography, Jinshan Hospital, Fudan University, Shanghai, China
| | - Xushuo Li
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Ying Yang
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai, China
| | - Ruixue Qi
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Shanghai, China,*Correspondence: Ruixue Qi,
| |
Collapse
|
28
|
Redmond WL, Kasiewicz MJ, Akporiaye ET. Enhancement of anti-tumor efficacy of immune checkpoint blockade by alpha-TEA. Front Immunol 2023; 14:1057702. [PMID: 36911733 PMCID: PMC9992800 DOI: 10.3389/fimmu.2023.1057702] [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: 09/29/2022] [Accepted: 02/10/2023] [Indexed: 02/24/2023] Open
Abstract
Cancer immunotherapy such as anti-PD-1/anti-PD-L1 immune checkpoint blockade (ICB) can provide significant clinical benefit in patients with advanced malignancies. However, most patients eventually develop progressive disease, thus necessitating additional therapeutic options. We have developed a novel agent, a-TEA-LS, that selectively induces tumor cell death while sparing healthy tissues, leading to increased activation of tumor-reactive T cells and tumor regression. In the current study, we explored the impact of combined a-TEA-LS + ICB in orthotopic and spontaneously arising murine models of mammary carcinoma. We found that a-TEA-LS + ICB led to increased production of pro-inflammatory cytokines that were associated with a reduction in tumor growth and prolonged survival. Together, these data demonstrate the potential utility of a-TEA-LS + ICB for the treatment of breast cancer and provide the rationale for clinical translation of this novel approach.
Collapse
Affiliation(s)
- William L Redmond
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, United States
| | - Melissa J Kasiewicz
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, United States
| | | |
Collapse
|
29
|
Mei T, Wang T, Deng Q, Gong Y. The safety of combining immune checkpoint inhibitors and platinum-based chemotherapy for the treatment of solid tumors: A systematic review and network meta-analysis. Front Immunol 2023; 14:1062679. [PMID: 36825025 PMCID: PMC9941623 DOI: 10.3389/fimmu.2023.1062679] [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: 10/06/2022] [Accepted: 01/23/2023] [Indexed: 02/10/2023] Open
Abstract
Objective Combination treatment regimens consisting of both immune checkpoint inhibitors (ICI) and chemotherapeutic agents have emerged as the standard of care for a range of cancers. This network meta-analysis (NMA) examined the toxicity profiles and safety rankings of these different ICI-based combination regimens. Methods The PubMed, EMBASE, Web of Science, and Cochrane Library databases were searched for all randomized controlled trials (RCTs) published as of March 1, 2022 comparing two or more treatment regimens in which at least one arm was comprised of an ICI + platinum-based chemotherapeutic regimen. Treatment-related adverse events (AEs) of any grade and AEs of grade 3 or higher were the primary endpoints for this analysis, while specific AE types were secondary endpoints. This NMA combined both direct and indirect comparisons when analyzing odds ratios (ORs) and the surface under the cumulative ranking curve (SUCRA) for different ICI-based treatment regimens. Results In total, 33 RCTs enrolling 19,012 cancer patients were included in this NMA. Of the analyzed regimens, avelumab + chemotherapy and camrelizumab + chemotherapy were associated with a significantly greater risk of AEs of any grade relative to ipilimumab + chemotherapy, durvalumab + chemotherapy, or pembrolizumab + chemotherapy. No significant differences in the risk of AEs of grade 3 or higher were observed when comparing different ICI regimens. Hepatotoxicity and pyrexia were the most common AEs associated with atezolizumab + chemotherapy treatment. Ipilimumab + chemotherapy was associated with a relatively higher risk of gastrointestinal and skin toxicity. Skin toxicity and hypothyroidism were the major AEs associated with nivolumab + chemotherapy. Fatigue and pneumonia were the most common AEs respectively associated with sugemalimab + chemotherapy and pembrolizumab + chemotherapy regimens. Conclusions Of the evaluated regimens, camrelizumab + chemotherapy and avelumab + chemotherapy were associated with significantly higher rates of AEs of any grade, whereas durvalumab and sintilimab were relatively safe PD-L1 and PD-1 inhibitors, respectively, when administered in combination with platinum-based chemotherapy. However, none of the evaluated ICI + chemotherapy regimens exhibited any differences with respect to the incidence of grade 3 or higher AEs, offering guidance that may be of value in routine clinical practice.
Collapse
Affiliation(s)
- Ting Mei
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ting Wang
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qianyue Deng
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Youling Gong
- Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
30
|
Huang J, Chen X, Xia B, Ma S. Advances in CT features and radiomics of checkpoint inhibitor-related pneumonitis: A short review. Front Immunol 2023; 14:1082980. [PMID: 36756121 PMCID: PMC9899831 DOI: 10.3389/fimmu.2023.1082980] [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: 10/28/2022] [Accepted: 01/09/2023] [Indexed: 01/24/2023] Open
Abstract
Checkpoint inhibitor-related pneumonitis (CIP) is a complication of immunotherapy for malignant tumors that severely limits the treatment cycles as well as endangers patients' health. The chest CT imaging features or typing of CIP and the application of radiomics will contribute to the precise prevention, early diagnosis and instant treatment of CIP. This article reviews the advances in the CT features and the application of radiomics in CIP.
Collapse
Affiliation(s)
- Jie Huang
- Department of Thoracic Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xueqin Chen
- Department of Thoracic Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing Xia
- Department of Thoracic Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shenglin Ma
- Department of Thoracic Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
31
|
Co-Targeting Luminal B Breast Cancer with S-Adenosylmethionine and Immune Checkpoint Inhibitor Reduces Primary Tumor Growth and Progression, and Metastasis to Lungs and Bone. Cancers (Basel) 2022; 15:cancers15010048. [PMID: 36612044 PMCID: PMC9818024 DOI: 10.3390/cancers15010048] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/16/2022] [Accepted: 12/17/2022] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BCa) is the most prevalent cancer in females and has a high rate of mortality, especially due to increased metastasis to skeletal and non-skeletal sites. Despite the marked clinical accomplishment of immune checkpoint inhibitor (CPI) therapy in patients with several cancers, it has had limited success in luminal subtypes of BCa. Accordingly, recent efforts have focused on combination therapy with CPI, including epigenetic modulators, to increase response rates of CPI in luminal BCa. We have previously shown that S-adenosylmethionine (SAM), the ubiquitous methyl donor, has strong anti-cancer effects in various cancers, including all subtypes of BCa. In the current study, we took a novel approach and examined the effect of CPI alone and in combination with SAM on tumor growth and metastasis in a syngeneic mouse model of luminal B BCa. We showed that SAM decreases cell proliferation, colony-formation (survival), and invasion of luminal B BCa cell lines (Eo771, R221A) in vitro. In in vivo studies, in Eo771 tumor-bearing mice, either SAM or anti-PD-1 antibody treatment alone significantly reduced tumor growth and progression, while the SAM+anti-PD-1 combination treatment had the highest anti-cancer efficacy of all groups. The SAM+anti-PD-1 combination reduced the percentage of animals with lung metastasis, as well as total metastatic lesion area, compared to control. Additionally, the SAM+anti-PD-1 combination significantly reduced the skeletal lesion area and protected tibial integrity to a greater extent than the monotherapies in an Eo771 bone metastasis model. Transcriptome analysis of Eo771 primary tumors revealed significant downregulation of pro-metastatic genes, including Matrix metalloproteinases (MMPs) and related pathways. On the other hand, CD8+ T cell infiltration, CD8+ T cell cytotoxicity (elevated granzymes), and immunostimulatory genes and pathways were significantly upregulated by the combination treatment. The results presented point to a combination of SAM with CPI as a possible treatment for luminal B BCa that should be tested in clinical studies.
Collapse
|
32
|
Perrotta F, Chino V, Allocca V, D’Agnano V, Bortolotto C, Bianco A, Corsico AG, Stella GM. Idiopathic pulmonary fibrosis and lung cancer: targeting the complexity of the pharmacological interconnection. Expert Rev Respir Med 2022; 16:1043-1055. [DOI: 10.1080/17476348.2022.2145948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Fabio Perrotta
- - Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80131, Napoli, Italy
- - U.O.C. Clinica Pneumologica “L. Vanvitelli”, A.O. dei Colli, Ospedale Monaldi, 80131, Napoli, Italy
| | - Vittorio Chino
- - University of Pavia Medical School, 27100 Pavia, Italy
- - Department of Medical Sciences and Infective Diseases, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, Pavia, Italy
| | - Valentino Allocca
- - Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80131, Napoli, Italy
- - U.O.C. Clinica Pneumologica “L. Vanvitelli”, A.O. dei Colli, Ospedale Monaldi, 80131, Napoli, Italy
| | - Vito D’Agnano
- - Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80131, Napoli, Italy
- - U.O.C. Clinica Pneumologica “L. Vanvitelli”, A.O. dei Colli, Ospedale Monaldi, 80131, Napoli, Italy
| | - Chandra Bortolotto
- - Dept. of Clinical-Surgical, Diagnostic and Paediatric Sciences, University of Pavia Medical School, Pavia, Italy
- - Department of Intensive Medicine, Unit of Radiology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Andrea Bianco
- - Department of Translational Medical Sciences, University of Campania “L. Vanvitelli”, 80131, Napoli, Italy
- - U.O.C. Clinica Pneumologica “L. Vanvitelli”, A.O. dei Colli, Ospedale Monaldi, 80131, Napoli, Italy
| | - Angelo Guido Corsico
- - Department of Medical Sciences and Infective Diseases, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, Pavia, Italy
- - Dept. of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
| | - Giulia Maria Stella
- - Department of Medical Sciences and Infective Diseases, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo Foundation and University of Pavia Medical School, Pavia, Italy
- - Dept. of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, Pavia, Italy
| |
Collapse
|
33
|
Rebuzzi SE, Prelaj A, Friedlaender A, Cortellini A, Addeo A, Genova C, Naqash AR, Auclin E, Mezquita L, Banna GL. Prognostic scores including peripheral blood-derived inflammatory indices in patients with advanced non-small-cell lung cancer treated with immune checkpoint inhibitors. Crit Rev Oncol Hematol 2022; 179:103806. [PMID: 36087850 DOI: 10.1016/j.critrevonc.2022.103806] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/28/2022] [Accepted: 09/05/2022] [Indexed: 10/31/2022] Open
Abstract
Peripheral blood inflammatory indices, like the neutrophil-to-lymphocyte ratio (NLR), may reflect the host's pro-inflammatory status and systemic immune response to cancer-related inflammation. We reviewed 22 combined prognostic scores based on peripheral blood-derived inflammatory indices for aNSCLC patients treated with single-agent or combination immune-checkpoint inhibitors (ICI) as first-line or subsequent therapy lines and attempted evidence strength assessment and scoring. The Lung Immune Prognostic Index (LIPI), consisting of derived NLR and LDH, was the most studied score with validated prognostic value in over five thousand aNSCLC ICI-naïve or pretreated patients. The combination of NLR and tumour programmed-cell-death-ligand1 (PD-L1) expression showed a predictive value. The Lung-Immune-Prognostic score (LIPS) might help identify patients with poor performance status but a favourable outcome following first-line ICI. These non-expensive scores can help clinicians discuss the prognosis with aNSCLC patients approaching ICI, identify those less likely to benefit from single-agent ICI and orient future clinical research.
Collapse
Affiliation(s)
- Sara Elena Rebuzzi
- Medical Oncology Unit, Ospedale San Paolo, Savona, Italy; Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, Genoa, Italy
| | - Arsela Prelaj
- Medical Oncology Department 1, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy; Department of Electronics, Information, and Bioengineering, Politecnico di Milano, Milan, Italy
| | - Alex Friedlaender
- Department of Oncology, Clinique Générale Beaulieu, Geneva, Switzerland; Department of Oncology, University Hospital of Geneva, Geneva, Switzerland
| | - Alessio Cortellini
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Alfredo Addeo
- Department of Oncology, University Hospital of Geneva, Geneva, Switzerland
| | - Carlo Genova
- Department of Internal Medicine and Medical Specialties (Di.M.I.), University of Genoa, Genoa, Italy; UOC Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Abdul Rafeh Naqash
- Medical Oncology/TSET Phase 1 Program, Stephenson Cancer Center, University of Oklahoma, Oklahoma City, USA
| | - Edouard Auclin
- Medical Oncology, Hôpital Européen Georges Pompidou, AP-HP, Université Paris Cité, Paris, France
| | - Laura Mezquita
- Medical Oncology Department, Hospital Clinic of Barcelona, Spain; Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Spain; Department of Medicine, University of Barcelona, Spain
| | | |
Collapse
|
34
|
Identification and Development of an Age-Related Classification and Signature to Predict Prognosis and Immune Landscape in Osteosarcoma. JOURNAL OF ONCOLOGY 2022; 2022:5040458. [PMID: 36276293 PMCID: PMC9581613 DOI: 10.1155/2022/5040458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 09/17/2022] [Indexed: 11/17/2022]
Abstract
Background. In childhood and adolescence, the prevailing bone tumor is osteosarcoma associated with frequent recurrence and lung metastasis. This research focused on predicting the survival and immune landscape of osteosarcoma by developing a prognostic signature and establishing aging-related genes (ARGs) subtypes. Methods. The training group comprised of the transcriptomic and associated clinical data of 84 patients with osteosarcoma accessed at the TARGET database and the validation group consisted of 53 patients from GSE21257. The aging-related subtypes were identified using unsupervised consensus clustering analysis. The ARG signature was developed utilizing multivariate Cox analysis and LASSO regression. The prognostic value was assessed using the univariate and multivariate Cox analyses, Kaplan-Meier plotter, time-dependent ROC curve, and nomogram. The functional enrichment analyses were performed by GSEA, GO, and KEGG analysis, while the ssGSEA, ESTIMATE, and CIBERSORT analyses were conducted to reveal the immune landscape in osteosarcoma. Results. The two clusters of osteosarcoma patients formed based on 543 ARGs, depicted a considerable difference in the tumor microenvironment, and the overall survival and immune cell infiltration rate varied as well. Among these, the selected 23 ARGs were utilized for the construction of an efficient predictive prognostic signature for the overall survival prediction. The testing in the validation group of osteosarcoma patients confirmed the status of the high-risk score as an independent indicator for poor prognosis, which was already identified as such using the univariate and multivariate Cox analyses. Furthermore, the ARG signature could distinguish different immune-related functions, infiltration status of immune cells, and tumor microenvironment, as well as predict the immunotherapy response of osteosarcoma patients. Conclusion. The aging-related subtypes were identified and a prognostic signature was developed in this research, which determined different prognoses and allowed for treatment of osteosarcoma patients to be tailored. Additionally, the immunotherapeutic response of individuals with osteosarcoma could also be predicted by the ARG signature.
Collapse
|
35
|
Chiang CH, Chiang CH, Peng CY, Hsia YP, See XY, Horng CS, Chang YC, Shen XE, Wang SS, Tsai TC, Chen YJ, Ma KSK, Chen BS, Luan YZ, Tay ST, Shen CH, Chung KC, Chiang CH, Peng CM. Efficacy of cationic amphiphilic antihistamines on outcomes of patients treated with immune checkpoint inhibitors. Eur J Cancer 2022; 174:1-9. [PMID: 35964360 DOI: 10.1016/j.ejca.2022.07.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/27/2022] [Accepted: 07/07/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Cationic amphiphilic antihistamines have been shown to improve patient outcomes in immunogenic tumours, but whether they can augment and improve response to immunotherapy is unknown. We aim to evaluate the effect of cationic amphiphilic antihistamines in patients receiving immune checkpoint inhibitors (ICIs). METHODS We conducted a retrospective propensity score-matched cohort study at two tertiary referral centres in Taiwan between January 2015 and December 2021. Patients who received desloratadine, cyproheptadine, and ebastine were classified as cationic amphiphilic antihistamine users. The primary outcome was overall survival, and the secondary outcomes were progression-free survival and clinical benefit rate. Patients treated with cationic amphiphilic antihistamines were matched to patients who received non-cationic amphiphilic antihistamines based on variables including age, cancer type, stage, and history of allergic diseases. RESULTS A total of 734 ICI-treated patients were included. After matching, 68 cationic amphiphilic antihistamine and non-cationic amphiphilic antihistamine users remained for analysis. Compared with non-cationic amphiphilic antihistamine users, patients who received cationic amphiphilic antihistamines had a significantly longer median overall survival (24.8 versus 10.4 months; Log-rank, p = 0.018) and progression-free survival (10.6 versus 4.93 months; Log-rank, p = 0.004). The use of cationic amphiphilic antihistamines was associated with an approximately 50% lower risk of all-cause mortality (HR, 0.55 [95% CI: 0.34-0.91]). Survival benefits were not seen in patients who received cationic amphiphilic antihistamines before immune checkpoint blockade. These survival benefits were observed regardless of the generation of cationic amphiphilic antihistamines. CONCLUSION The use of cationic amphiphilic antihistamines was associated with improved survival among patients treated with immunotherapy.
Collapse
Affiliation(s)
- Cho-Han Chiang
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Boston, MA, USA
| | - Cho-Hung Chiang
- Division of General Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan; Division of Hematology and Oncology, Department of Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan; Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| | - Chun-Yu Peng
- Da Vinci Minimally Invasive Surgery Center, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yuan Ping Hsia
- Department of Family Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Xin Ya See
- Da Vinci Minimally Invasive Surgery Center, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chuan-Sheng Horng
- Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yu-Cheng Chang
- Da Vinci Minimally Invasive Surgery Center, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Xuan-Er Shen
- Da Vinci Minimally Invasive Surgery Center, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shih-Syuan Wang
- Da Vinci Minimally Invasive Surgery Center, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Tien-Chi Tsai
- Da Vinci Minimally Invasive Surgery Center, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Yuan-Jen Chen
- Da Vinci Minimally Invasive Surgery Center, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Kevin Sheng-Kai Ma
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Graduate Institute of Biomedical Electronics and Bioinformatics, College of Electrical Engineering and Computer, National Taiwan University, Taipei, Taiwan; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Yu-Ze Luan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Soon-Tzeh Tay
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Chin-Hsuan Shen
- School of Medicine, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Katharine Ching Chung
- Da Vinci Minimally Invasive Surgery Center, Chung Shan Medical University Hospital, Taichung, Taiwan
| | | | - Cheng-Ming Peng
- Da Vinci Minimally Invasive Surgery Center, Chung Shan Medical University Hospital, Taichung, Taiwan; School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| |
Collapse
|
36
|
A Ferroptosis-Related Gene Signature for Overall Survival Prediction and Immune Infiltration in Lung Squamous Cell Carcinoma. Biosci Rep 2022; 42:231598. [PMID: 35866375 PMCID: PMC9434561 DOI: 10.1042/bsr20212835] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/28/2022] [Accepted: 07/20/2022] [Indexed: 02/05/2023] Open
Abstract
Background: Ferroptosis is associated with cancer initiation and progression. However, the molecular mechanism and prognostic value of ferroptosis-related genes in lung squamous cell carcinoma (LUSC) are poorly understood. Methods: The mRNA expression profiles, methylation data, and clinical information of patients with LUSC were downloaded from TCGA and GEO database. Ferroptosis-related differentially expressed genes (DEGs) were identified between cancerous and non-cancerous tissues, and their prognostic value was systemically investigated by bioinformatic analyses. Results: A ferroptosis-related gene signature (ALOX5, TFRC, PHKG2, FADS2, NOX1) was constructed using multivariate Cox regression analysis and represented as a risk score. Overall survival (OS) probability was significantly lower in the high-risk group than in the low-risk group (P<0.001), and receiver operating characteristic curve showed a good predictive capacity (AUC = 0.739). The risk score was an independent prognostic factor for LUSC. A nomogram was constructed to predict the OS probabilities at 1, 3, and 5 years. High-risk score was associated with increased immune infiltration, lower methylation levels, higher immune checkpoint genes expression levels, and better chemotherapy response. Cell adhesion molecules, focal adhesion, and extracellular matrix receptor interaction were the main pathways in the high-risk group. The signature was validated using the TCGA test cohort, entire TCGA cohort, GSE30219, GSE157010, GSE73403, and GSE4573 datasets. The gene disorders in patients with LUSC were validated using real-time PCR and single-cell RNA sequencing analysis. Conclusions: A ferroptosis-related gene signature was constructed to predict OS probability in LUSC. This could facilitate novel therapeutic methods and guide individualized therapy.
Collapse
|
37
|
Ren W, Ren J, Zhang N, Liu X, Deng Y, Jiang Y, Yan B, Xiao X, Yu H. CBX7 represses the POU2F2 to inhibit the PD-L1 expression and regulate the immune response in bladder cancer. Biochem Biophys Res Commun 2022; 613:12-18. [DOI: 10.1016/j.bbrc.2022.04.114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 12/25/2022]
|
38
|
Treatment Sequences in Patients with Recurrent or Metastatic Head and Neck Squamous Cell Carcinoma: Cetuximab Followed by Immunotherapy or Vice Versa. Cancers (Basel) 2022; 14:cancers14102351. [PMID: 35625956 PMCID: PMC9139601 DOI: 10.3390/cancers14102351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The prognosis was poor when patients had recurrent or metastatic head and neck squamous cell carcinoma (R/M HNSCC). Herein, we conducted an observational study of cetuximab followed by immunotherapy (Cet-IO) versus immunotherapy followed by cetuximab (IO-Cet) in patients with R/M HNSCC. METHODS Patients who were diagnosed with R/M HNSCC and treated with a sequential cetuximab-containing regimen and immunotherapy-containing regimen were enrolled in our study. Kaplan-Meier curves were estimated for progression-free survival (PFS) and overall survival (OS). RESULTS A total of 75 patients were enrolled in our study for oncologic outcomes evaluation, with 40 patients in Cet-IO and 35 patients in IO-Cet. The median PFS1 was 5.1 months in Cet-IO and 4.5 months in IO-Cet (p = 0.777) and the median PFS2 was 16.5 months in Cet-IO and 11.4 months in IO-Cet (p = 0.566). The median OS was 23.7 months versus 22.8 months in Cet-IO and IO-Cet, respectively (p = 0.484). The overall response rate (ORR) were 73% in Cet-IO versus 37% in IO-Cet (p = 0.002). Multivariate analysis demonstrated that the treatment sequences, Cet-IO or IO-Cet, were insignificantly different with survival. CONCLUSION Both Cet-IO and IO-Cet are effective in R/M HNSCC patients with insignificant survival differences. The higher ORR of Cet-IO might render it to be considered in patients with large tumor burdens and urgent needs for treatment responses. Further prospective studies are merited to validate our conclusions.
Collapse
|
39
|
Imyanitov EN, Iyevleva AG. Molecular tests for prediction of tumor sensitivity to cytotoxic drugs. Cancer Lett 2022; 526:41-52. [PMID: 34808283 DOI: 10.1016/j.canlet.2021.11.021] [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: 09/25/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 11/15/2022]
Abstract
Chemotherapy constitutes the backbone of cancer treatment. Several predictive assays assist personalized administration of cytotoxic drugs and are recommended for use in a clinical setting. The deficiency of DNA repair by homologous recombination (HRD), which is caused by inactivation of BRCA1/2 genes or other genetic events, is associated with high tumor responsiveness to platinum compounds, bifunctional alkylating agents and topoisomerase II poisons. Low activity of MGMT predicts the efficacy of nitrosoureas and tetrazines. Some clinically established pharmacogenetic tests allow for the adjustment of drug dosage, for example, the analysis of DPYD allelic variants for administration of fluoropyrimidines and UGT1A1 genotyping for the use of irinotecan. While there are promising molecular predictors of tumor sensitivity to pemetrexed, gemcitabine and taxanes, they remain in the investigational stage and require additional validation. Comprehensive molecular analysis of tumors obtained from drug responders and non-responders is likely to reveal new clinically useful predictive markers for cytotoxic therapy.
Collapse
Affiliation(s)
- Evgeny N Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, 197758, Russia; Department of Medical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg, 194100, Russia; Department of Oncology, I.I. Mechnikov North-Western Medical University, St.-Petersburg, 191015, Russia.
| | - Aglaya G Iyevleva
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, 197758, Russia; Department of Medical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg, 194100, Russia
| |
Collapse
|